Monday, December 5, 2011

Brain Plasticity and Disease: AMatter of Inhibition: A Review Article

Interesting article by Laura Baroncelli and Colleagues.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

A role for glia in the progression of Rett’s syndrome

Interesting article by Daniel T. Lioy.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

INSIDE THE MINDS OF MICE AND MEN

Interesting article by MONYA BAKER.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

Subclinical myocardial dysfunction in Rett syndrome

Interesting article by Claudio De Felice and colleagues.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

MeCP2 and Rett syndrome: reversibility and potential avenues for therapy: A Review Article

Interesting article by Kamal K.E. GADALLA and colleagues.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

Folinic Acid supplementation in Rett syndrome patients does not influence the course of Disease: A Randomized Study

Interesting article by Eveline E. O. Hagebeuk and colleagues.

Note: Please click on the title to read the full article.

Source: NCBI-PubMed

Movement Disorders Emergencies in Childhood

A very Interesting article by F.J. Kirkham and Colleagues.


Note: Click on the title to read the full article.

Source:NCBI-Pubmed

Rett Syndrome: Exploring the Autism Link

A very Interesting article by Dr. Alan K Percy.

Must read.

Note: Click on the title to read the full article.

Source:NCBI-Pubmed

Tuesday, November 8, 2011

Rett Syndrome Research Trust Launches Awareness Campaign in Iconic Times Square Location

Trumbull, CT (PRWEB) November 03, 2011

The Rett Syndrome Research Trust is pleased to announce a campaign to boost awareness of Rett Syndrome in the most iconic of all advertising venues – Times Square in New York City. A newly created public service announcement will run an average of nine times an hour for three months starting November 1st on the colossal 6000 square foot Mediamesh display at the corner of 42nd Street and 8th Avenue. An estimated 1.5 million people are expected to view the PSA daily.

Rett Syndrome strikes little girls almost exclusively, with first symptoms usually appearing before the age of 18 months. These children lose speech, motor control and functional hand use, and many suffer from seizures, orthopedic and severe digestive problems, breathing and other autonomic impairments. Most live into adulthood and require total, round-the-clock care. Rett Syndrome is caused by mutations in a gene called MECP2.

The Rett Syndrome Research Trust is the premier organization devoted exclusively to promoting international research on Rett Syndrome and related MECP2 disorders.

While Rett Syndrome has become a high-profile disorder in scientific circles, it is still fairly unknown in the lay and medical communities; thousands of girls and women remain un- or misdiagnosed. We welcome this unique opportunity to help raise the profile of this disease, which has been dramatically reversed in pre-clinical models.

“I am so thankful to Bill Koenigsberg, CEO and Founder of Horizon Media, who secured this spectacular advertising real estate; Garage Media, owner of the Port Authority Display; A2AMedia, who manages the screen, for giving us the opportunity to showcase our disorder; and Jon Denny, film and TV producer, for creating our PSA. When smart, talented, generous people come together for a good cause, amazing things can happen,” said Monica Coenraads, Executive Director of RSRT and parent of a teenaged daughter severely disabled by Rett Syndrome.

“We are thrilled to help RSRT get out their message for such a worthy cause,” said Bill Koenigsberg.

About the Rett Syndrome Research Trust
The Rett Syndrome Research Trust is the premier organization devoted exclusively to promoting international research on Rett Syndrome and related MECP2 disorders. Our goal is clear: to heal children and adults who will otherwise suffer from this disorder for the rest of their lives. With our experience and tight focus, RSRT has an unparalleled knowledge base and extensive networking abilities in the world of high-level research. This puts RSRT in a unique position to stimulate, evaluate, support and monitor ambitious and novel scientific projects. To learn more about the Trust, please visit http://www.ReverseRett.org

Source: PRWEB

A Mid-Michigan Family Shares Their Experience With Rett Syndrome

Source: WLNS.com

Note: Click on the title to read the full news.

Families Raise Awareness About Rett Syndrome

By DAN MANNARINO Staff reporter

12:23 a.m. EDT, October 16, 2011
NEW YORK (PIX11)—
Imagine you have thoughts, hopes, and dreams but no way to actually express them. It's a rare disorder called Rett Syndrome that affects young girls. Worldwide, 20 girls are born every day with the disorder.

Dr. Sasha Djukic says the disorder doesn't begin right at birth. "Imagine you have a child who is 1 year old, happy and healthy, and then the disease starts, in a matter of a few months it drops the ability to speak, use her hands, walk," says Djukic.

Abby Diamond is now a beautiful, 8-year-old girl. At age 3, she was diagnosed with Rett Syndrome and stopped walking and communicating with her family.

The problem is, not many people know about the disease, so not many people donate money to research.

So today, under perfectly blue skies, families gathered on the steps of the Tweed Courthouse in Lower Manhattan and in other locations around the globe to raise awareness about Rett Syndrome.

"Climbing steps is a symbolic gesture, bringing you upward and forward," says Djurik.

In an inspiring sight to watch, the girls slowly but surely climbed the 30 steps. Joining them were members of the New York City Fire Department who, in some cases, carried these brave girls to the top.

"The girls are good girls, pure in nature, friendly and trapped inside these bodies," says Abby's father, Erick Diamond.

CAL artists support Rett Syndrome fundraiser

Thursday, August 25, 2011

A ray of hope for Indian Rett Angels and their families

Renowned neuro-scientist, Dr Mriganka Sur is all set to collaborate with the All India Institute of Medical Sciences (AIIMS), New Delhi for a research on finding a cure for neuro development diseases like Autism, Rett syndrome, etc.

Speaking to the Chandigarh Newsline, Dr Sur, professor of Neurosciences and head, department of Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), USA, said, “We are planning to collaborate on a research programme on neuro development disorders. Despite the rarity of the disease, Rett syndrome which is found 95 per cent in girls due to their mutations, in India, the percentage of girls suffering from it is quite high. Even in USA, one in every 150 children is prone to Autism and in the United Kingdom, the chances of having autism are even higher, one in every 100. Though there are clinical trials and constant researches going on, there is no cure yet for most neuro development disorders.”

Dr Sur, who is the director of Simon’s Initiative on Autism, said, “Though some studies have established that Autism is mostly a genetic disease, yet gene therapy is no cure for this genetic disorder.”

Though there are no pre natal tests available to detect if the child could be born with autism, however, Dr Sur warns that autism in family history may be a sign. “If anyone in the family has a history of autism, chances are that the baby could be born with the disorder too. But instead of losing hope, would be parents should approach genetic counsellors or informed physicians.”

The available therapies for autism include applied behaviour analysis (ABA), developmental models, structured teaching, speech and language therapy, social skills therapy, and occupational therapy, he added.

The diseases

Autism spectrum disorder (ASD) is a range of complex neuro development disorders, characterised by social impairments, communication difficulties, and restricted, repetitive, and stereotyped patterns of behaviour. Usually, the symptoms start showing before the child is three years old.

Rett syndrome is a childhood neuro developmental disorder that affects females almost exclusively. The child generally appears to grow and develop normally, before symptoms begin. Early symptoms may include loss of muscle tone, a slowing of development, problems crawling or walking, and diminished eye contact. As the syndrome progresses, a child will lose purposeful use of hands and the ability to speak. The inability to perform motor functions is perhaps the most severely disabling feature of Rett syndrome, interfering with body movement.

Source: Indian express.com

Note: To read the original post please click on the title of this news.

Thursday, August 4, 2011

Fourth Annual Rett syndrome Meeting and Symposium: 23rd October 2011

Dear All,

As October is "Rett syndrome Awareness Month", So Indian Rett Syndrome Foundation is going to organize the Fourth Annual Rett Syndrome Awareness Meet/Symposium with help of Department of Pediatrics, AIIMS, New Delhi.

This meeting will be joined by Parents from different parts of India and Healthcare professionals (Doctors, Therapists, Scientists and Researchers). This one day Meeting/symposium will include lectures on the basics of Rett syndrome and its management, Parent-Doctor interactive sessions and therapy sessions.

Come and Join us to educate yourself and raise awareness about Rett Syndrome, a neurodevelopmental disorder. which primarily affects girls known as "Silent Angels".

We are also looking forward for sponsorships for this event and donations for this cause. Your help and support can improve lives of these little Angels and their families.

Date of Event: 23rd October, 2011
Day: Sunday
Time: 9.30am
Venue: Conference Hall, First Floor, JLN Auditorium,
All India Institute of Medical Sciences
Ansari Nagar
New Delhi-110029


Please save the date in your calenders and let your family/friends know about this by spreading the word.

If you have any queries in regard of this event, please contact us at

info@rettsyndrome.in, info.rett@yahoo.com

Registration to this event is totally free but limited. For registration for this event, please contact

rkhajuria@rettsyndrome.in
admin@rettsyndrome.in

Regards,

Indian Rett Syndrome Foundation
12/1, sector –1, Pushp Vihar,
New Delhii -110017
INDIA
Website: www.rettsyndrome.in
E-mail: info@rettsyndrome.in, admin@rettsyndrome.in, info.rett@yahoo.com
Blog: www.rettsyndromeindia.blog​spot.com
Ph# +919999343421

Tuesday, June 28, 2011

Ex Vivo Treatment with a Novel Synthetic Aminoglycoside NB54 in Primary Fibroblasts from Rett Syndrome Patients Suppresses MECP2 Nonsense Mutations

A recent and very interesting article by Vecsler and colleagues

Abstract

BACKGROUND:

Nonsense mutations in the X-linked methyl CpG-binding protein 2 (MECP2) comprise a significant proportion of causative MECP2 mutations in Rett syndrome (RTT). Naturally occurring aminoglycosides, such as gentamicin, have been shown to enable partial suppression of nonsense mutations related to several human genetic disorders, however, their clinical applicability has been compromised by parallel findings of severe toxic effects. Recently developed synthetic NB aminoglycosides have demonstrated significantly improved effects compared to gentamicin evident in substantially higher suppression and reduced acute toxicity in vitro.

RESULTS:

We performed comparative study of suppression effects of the novel NB54 and gentamicin on three MECP2 nonsense mutations (R294X, R270X and R168X) common in RTT, using ex vivo treatment of primary fibroblasts from RTT patients harboring these mutations and testing for the C-terminal containing full-length MeCP2. We observed that NB54 induces dose-dependent suppression of MECP2 nonsense mutations more efficiently than gentamicin, which was evident at concentrations as low as 50 µg/ml. NB54 read-through activity was mutation specific, with maximal full-length MeCP2 recovery in R168X (38%), R270X (27%) and R294X (18%). In addition, the recovered MeCP2 was translocated to the cell nucleus and moreover led to parallel increase in one of the most important MeCP2 downstream effectors, the brain derived neurotrophic factor (BDNF).

CONCLUSION:

Our findings suggest that NB54 may induce restoration of the potentially functional MeCP2 in primary RTT fibroblasts and encourage further studies of NB54 and other rationally designed aminoglycoside derivatives as potential therapeutic agents for nonsense MECP2 mutations in RTT.

Note: Click on the title to read the full article


Source: NCBI-Pubmed

Tuesday, May 31, 2011

Physical and Mental Health of Mothers Caring for a Child With Rett Syndrome

By

Crystal L. Laurvick, MPH, Michael E. Msall, MD,Sven Silburn, MS (ClinPsych) Carol Bower, PhD, Nicholas de Klerk, PhD,Helen Leonard, MBChB

ABSTRACT

OBJECTIVES. Our goal was to investigate the physical and mental health of mothers who care for a child with Rett syndrome.

METHODS.

We assessed maternal physical and mental health by using the SF-12 version 1 physical component summary and mental component summary scores as the outcome measures of interest. Mothers (n 135) of children with Rett syndrome completed the SF-12 measure as part of the Australian Rett Syndrome Study in 2002. The analysis investigated linear relationships between physical and mental health scores and maternal, family, and child characteristics.

RESULTS. Mothers ranged in age from 21 to 60 years and their children from 3 to 27years. Nearly half of these mothers (47.4%) indicated that they worked full-time or part-time outside the home, and 41% had a combined family (gross) income of 40 000 Australian dollars. The resultant model for physical health demonstrated that the following factors were positively associated with better maternal physical health: the mother working full-time or part-time outside the home, having some high school education, having private health insurance, the child not having breathing problems in the last 2 years, the child not having home-based structured therapy, and high scores on the Family Resource Scale (indicating adequacy of time resources for basic and family needs). The resultant model for mental health demonstrated that the following factors were positively associated with better maternal mental health: the mother working full-time or part-time outside the home, the child not having a fracture in the last 2 years, lesser reporting of facial stereotypes and involuntary facial movements, being in a well-adjusted marriage, and having low stress scores.

CONCLUSIONS. Our study suggests that the most important predictors of maternal physical and emotional health are child behavior, caregiver demands, and family function.

Note: Click on the title to read the full article.

Source: Pubmed

Monday, May 30, 2011

Daily tips for day to day living with Rett syndrome : Advice from the Internation Rett Syndrome Foundation Experts

Bathing

If she does not sit without support, it may be hard to get her in and out of the tub and keep her stable and safe while you wash her.

There are a number of waterproof bath chairs available, ranging from mesh sling-back styles to rigid plastic benches and chairs.

A hand-held shower hose can be very useful in the tub, especially for hair washing.

Waterproof wheelchairs are available for use in roll-in showers.

A mesh table that hooks into the back of a roll-in shower provides a safe way to bathe without undue bending by the caregiver.

Lifting to Prevent Back Injuries

It is important to learn how to lift correctly, so that you place the least amount of strain on your body, particularly your back. It is a good idea to attend one of your daughter’s PT sessions to get suggestions for the best ways to care for her with the least lifting possible. The school district may even allow you to have special PT sessions for training purposes. Ask your school administrator or request it in her IEP. While good lifting techniques can prevent injuries, they will also make transfers safer and easier. A training session should include: transferring in and out of your car, in and out of the tub, in and out of sitting to stand position, placing the wheelchair in and out of the car, and any other situations which pull on your back. When lifting, widen the base of your stance and bend your legs while keeping your back straight. When transferring from bed to chair, lean your daughter forward into your shoulder. A sliding board is an inexpensive piece of equipment which can be used for safe transfers. If your daughter is able to learn to assist you, practice often so she can gain strength to help. There are a number of mechanical devices when lifting becomes too difficult. Ramps, platform lifts, stairway lifts, and elevators increase accessibility and decrease back strain. Bathtub and bedside lifts can be invaluable. When changing diapers, it is better to roll the girl on her side to place the diaper under her rather than to lift her hips. It may not seem like a lot of strain, but over time, lifting the wrong way can be harmful.

The general principles of lifting are:

* the force is against you
* lift from the waist
* add upper body weight
* watch your weight
* warm up before lifting


The causes of back injuries include:

* lifting from the waist, and twisting
* lifting overhead or away from you
* heavy, repetitive lifting
* awkward position
* unbalanced load
* sitting/standing in one position too long

Traveling

Vacation and Travel For Families with Special Needs Children:

Most families with children choose to vacation and travel during the summer months, and it may take some planning to have a successful vacation. Even more so for families with children who have special needs. There are many things to consider when planning a vacation: destination, transportation, hotel or other overnight arrangements, activities, adaptations, and general accessibility, to name a few.

Tips for Air Travel:

* Contact the airline as early as possible for special assistance, or use a travel agent who is familiar with making special reservations.
* If possible, ask for a non-stop flight to avoid airport shuffle.
* If you use a car seat, make sure it is certified for air travel.
* Make sure to label all equipment with your name, address, and phone number.
* If your child’s wheel chair is checked, remind the flight attendant before landing so she can make a radio inquiry to make sure the chair is delivered promptly.
* Make arrangements for special meals in advance. Do take along some of your child’s favorite snacks and drinks.
* Take your child to the toilet or change her diaper before the flight.
* Invest in a Walkman or DVD player and a good set of barrettes to anchor the headset.
* Offer your child something to eat or drink during descent.

Additional Travel Tips:

* Make advanced arrangements with car rental services.
* Before you leave home, get a disabled parking placard that can be used everywhere.
* Check with your hotel to be sure your room and facilities are handicap accessible.
* The following is a list of Web sites, which addresses those considerations. They contain information about vacation travel through articles, books, Internet links, and other resources.

1. A wonderful Directory of Summer Camps for Children with Disabilities was compiled by the National Information Center for Children and Youth with Disabilities (NICHCY). It contains disability-specific as well as general camp resources.
2. The Global Access Disabled Travel Network is a Web site containing personal travel experiences, as well as information on travel books, specific destinations, tips on planning, hotels and resorts, etc.
3. Travel with Kids offers information and links to cruise ship accessibility, tour operators and travel agents specializing in special needs, outdoor vacations for people with disabilities, and much more.
4. The Parenting Special Needs Web site has a section on Accessible Summer Recreation. It includes accessible travel, camps for special children, recreation and sport, adaptive equipment, and also just for fun activities for children with special needs.
5. The Enabled RVer hosts articles on topics related to RV travel, such as accessible RVs, the Handicapped Travel Club (HTC), travel guides, campgrounds and specific destinations, resources, and adaptive equipment.
6. Access-Able Travel Source contains information on travel with disabilities, mature travel, disability magazines, access guides, wheelchair travel, scooter rental, accessible transportation, world destinations, lists of travel professionals, links, and tips for the traveler with disabilities.
7. The Project ACTION Accessible Travelers' Database was created to assist tourists with disabilities to access mass transit systems while traveling to other cities. The database includes information on hotel/motel shuttles, accessible taxis, private bus/tour companies, van rentals, public transit operators, and national 800 numbers.
8. The Disability Travel and Recreation Resources web site includes information and links on topics such as travel planning, destinations, transportation, air travel, children, and books.
9. Emerging Horizons is a magazine, available online and in print, about accessible travel for people with mobility disabilities. It includes access information, resources, news and travel tips.
10. The Society for Accessible Travel & Hospitality (SATH) actively promotes awareness, respect, accessibility, and employment for persons with disabilities in the tourism industry. SATH contains disability-specific 'How To Travel' articles, as well as an extensive list of resources for the traveler with disabilities.
11. Accessible Recreation on Federal Lands provides a listing of government resources that manage federal recreation sites in the United States. It also includes the Accessibility Data Management System (ADMS), an online resource containing accessibility information about many federal facilities.
12. The National Center on Physical Activity and Disability web site contains a virtual library with an extensive list of resource directories, fact sheets, bibliographies, and monographs. Topics range from adaptive equipment, adapting activities in recreation programs, camping, hiking, fishing, scuba, and much more.

Although the sources listed above seem extensive, it is just the tip of the iceberg. Explore what will best fit you and your needs as a family

Source: International Rett Syndrome Foundation (www.rettsyndrome.org)

Monday, May 23, 2011

Longevity in Rett Syndrome: Analysis of the North American Database

Interesting article by

Russell S. Kirby and colleagues



Source: NCBI-Pubmed


Note: Click on the title post to read the full article

Long-term follow-up of functioning after spinal surgery in patients with Rett syndrome

Interesting article by

Eva-Lena Larsson and colleagues


Source: NCBI-Pubmed

Note: Click on the title post to read the full article

Autism Spectrum Disorders

Judith H Miles, MD, PhD
Thompson Center for Autism and Neurodevelopmental Disorders & Department of Child Health
University of Missouri Hospitals and Clinics
Columbia, MO
milesjh@missouri.edu

Rebecca B McCathren, PhD
Early Childhood Special Education
College of Edicuation
University of Missouri
Columbia, MO
mccathrenr@missouri.edu

Janine Stichter, PhD
Thompson Center for Autism and Neurodevelopmental Disorders & Department of Special Education
University of Missouri
Columbia, MO
stichterj@missouri.edu

Marwan Shinawi, MD
Department of Pediatrics, Division of Genetics and Genomic Medicine
Washington University School of Medicine
St. Louis, MO
Shinawi_M@kids.wustl.edu

Summary

Disease characteristics. Autism comprises a clinically heterogeneous group of disorders – collectively referred to as “autism spectrum disorders” (ASD) – that share common features of impaired social relationships, impaired language and communication, and repetitive behaviors or a narrow range of interests. For most children with autism, symptoms develop gradually, although approximately 30% have a "regressive" onset usually between ages 18 and 24 months. About 50%-70% of children with autism are identified as intellectually disabled by nonverbal IQ testing and approximately 25% develop seizures. Autism can be considered complex (i.e., presence of dysmorphic features and/or microcephaly) or essential (i.e., absence of physical abnormalities and microcephaly). About 25% of children who fit the diagnostic criteria for ASD at age two to three years subsequently begin to talk and communicate, and by age six to seven years blend to varying degrees into the regular school population. The remaining 75% have lifelong disability requiring intensive parental, school, and social support.

Diagnosis/testing. The behavioral criteria presented in the American Psychiatric Association Manual of Psychiatric Diseases, 4th edition (DSM-IV) remain the standard for making an autism diagnosis in the US. Currently, three subgroups (autistic disorder, Asperger syndrome, and PDD-NOS) are recognized. To qualify for a diagnosis of autistic disorder (i.e., classic autism), a child must have shown abnormalities in social interaction and language used for social communication or symbolic/imaginative play and either stereotypic motor mannerisms or restricted patterns of interest before age three years. If the child does not meet criteria for autistic disorder, he or she may be given a diagnosis of Asperger syndrome (AS) or pervasive developmental disorder-not otherwise specified (PDD-NOS), the other two disorders included in ASD. Autism has many etiologies, a concept that was widely embraced following the discovery of the molecular basis of Rett syndrome in 2006. Since then autism has been documented in hundreds of neurologically based syndromes with multiple causes, outcomes, and treatment responses. Currently, an etiology can be identified for between 15% and 20% of individuals with autism; in the others the cause remains unknown.

Management. Treatment of manifestations: Management of autism involves educational, behavioral, and medical therapies to promote conversational language and social interactions while mitigating repetitive self-stimulatory behaviors, tantrums, aggression, and self-injurious behaviors. The mainstay of therapy is early individualized intensive training either in the school or home, where the environment must be predictable with planned transitions between activities and venues. Visual supports are helpful in promoting language acquisition. The Visually Cued Instruction and Schedules program uses graphic clues to aid communication, organizational skills, and self-management. The Picture Exchange Communication System provides a visual system in which to learn communication. Social Stories intervention increases appropriate behavior by explaining social situations in ways understandable to the student. Medications, especially atypical antipsychotics, can ameliorate specific symptoms such as aggressive or self-injurious behavior. Children treated early can usually be taught, to varying degrees, to communicate, recognize and respond to social interactions, develop imaginative play, and curb all-consuming repetitive self-stimulatory behaviors.

Prevention of secondary complications: Intensive behavioral intervention programs can prevent or at least mitigate aggressive behaviors (tantrums, aggression, and self injurious behaviors) that occur commonly in children with no communication skills or interest in social interactions.

Surveillance: At least annual surveillance for health, developmental progress, education milestones, behavioral and family functioning preferably at a multidisciplinary autism clinic is strongly recommended.

Testing of relatives at risk: Because sibs are at increased risk of developing autism or an ASD, their language, development, and behavior should be closely monitored for the first three years, preferably through an autism sib program.

Genetic counseling. For individuals with autism in whom the etiology is known, genetic counseling and risk assessment are based on the genetics of that specific diagnosis. For autism of unknown cause, the empiric aggregate risk to sibs is 5%-10% for autism and 10%-15% for milder conditions, including language, social, and psychiatric disorders. For families with two or more affected children, the recurrence risk approaches 35%. Male sibs (brothers) of a proband with essential autism have a 7% risk for autism and an additional 7% risk for milder ASD. Female sibs (sisters) of a proband with essential autism have a 1% risk for autism; the risk for a milder ASD is unknown. The recurrence risk to sibs of a proband with complex autism is 1% for autism and an additional 2% for a milder ASD.

Definition

Clinical Manifestations

Autism spectrum disorders (ASD) develop prior to age three years. Infants typically do not care to be held or cuddled and do not reach out to be picked up. Often they are "colicky" and hard to console, typically quieting more readily when left alone. They may avoid and fail to initiate eye contact or stare into space. Sleep disturbances and sensory issues may be noted in the first year. Despite early signs, children with ASD often do not come to medical attention until after the second year when language delays are obvious.

For most children, the onset of ASD symptoms is gradual; however, approximately 30% have a "regressive" onset. These children begin to speak and then, either gradually or precipitously, lose language and become distant. Within a matter of days, the child may refuse to make eye contact and stop responding to his/her name. Deafness is often suspected, although hearing tests are normal. Repetitive movements may develop immediately or not until the child is age three or four years. Though it has been debated whether these children are well and then become damaged by some exogenous exposure, the best evidence, including retrospective analysis of first birthday videotapes and neuropathologic studies, suggests their regressive course is genetically determined [Lord et al 2004, Volkmar et al 2005, Werner & Dawson 2005, Stefanatos 2008].

Approximately 25% of children who fit the diagnostic criteria for ASD at age two or three years subsequently begin to talk and communicate and by age six or seven years blend to varying degrees into the regular school population. Even for this group, social impairments generally continue. For the remaining 75%, most have some improvement with age but continue to require parent, school, and societal support. Excellent reviews of outcomes are provided by Howlin et al [2000], Howlin et al [2004], Seltzer et al [2004], and Farley et al [2009]. Some studies indicate that fewer than 5% of children with autism completely recover [Nordin & Gillberg 1998]; however, loosening of diagnostic criteria to include less impaired children appears to be increasing that number. A 20-year follow-up of adults between ages 22 and 46 years diagnosed with autism and average or near-average cognitive abilities in the 1980s found that half the individuals functioned quite well and half were employed in full-time or part-time paying jobs; however, only 12% lived independently and 56% lived with their parents [Farley et al 2009].

Autism spectrum disorders are defined completely on the basis of three areas of behavioral impairment:

*

Impairments in social interaction. Impairment in social interaction distinguishes individuals with autism from those around them. Children with classic autism are unable to "read" other people, ignoring them and often strenuously avoiding eye contact. Typically, they do not comfort others or seek comfort and do not share interests with others, such as bringing toys or pictures to their parents. Rather, they use their parents as objects, and may climb on them to get to a desired object, pull the parent by the hand, or place the parent's hand on the object, as if the child were using a tool. In clinic, the child who is content to turn pages of a magazine or spin the wheels of a car may become agitated when a simple examination is attempted. At home, the child with autism usually prefers to be by himself, engaging in his own, often repetitive, activities. The lack of functional or spontaneous make-believe play is characteristic. Toys are lined up, sorted, twirled, or hurled, but are not used for imaginative games or imitation of day-to-day activities, such as feeding the baby or washing the dishes. When play emerges later, it is stylized and not spontaneous. Children with autism fail to develop friendships with peers and siblings. In school, they often stand and watch other children from a distance. Some children respond to social overtures but take little social initiative, while others seek interaction but have little sense of how to proceed toward normal friendships.
*

Impairments in communication. Children with autism fail to develop reciprocal communication either by speech, gestures, or facial expressions. Characteristically, young children fail to use eye gaze or pointing to communicate and direct their parent’s attention. Early pragmatic skills are limited and are characterized by typical rates of requesting but substantially reduced rates of social interaction and establishing joint attention. Deficits in pragmatic skills are present throughout life and affect both language and social interaction. The young child appears unable to grasp the concept that speech can be used to name objects, to request a toy, or to engage others. In contrast to the child with nonspecific intellectual disability or a primary developmental language disorder who usually has better receptive than expressive language, the child with autism has impaired receptive language. When children with autism learn to talk, they display stereotypic speech that may involve echolalia, pronoun reversal, and unusual inflections and intonations. Unlike typically developing children who begin talking using one-word utterances, children with autism may begin talking in "chunks" composed of commercials, movies, or others' speech. These chunks often convey idiosyncratic meanings and the child with autism has no understanding of the conventional meaning of the individual words. Pragmatic difficulties including difficulties sustaining a conversation, turn taking, and allowing the conversational partners to introduce their topics, usually continue despite improvement in expressive speech. [Lord et al 2004].
*

Repetitive and stereotypic behaviors. Infants may stare or rock. Toddlers may have motor "stereotypies" such as movements of fingers, twirling strings, flicking pages of books, or licking. Repetitive whole body movements may include spinning and running back and forth. The repetitive behaviors often have a visual component such as holding the fingers to the side of the face and watching them with a sideways glance. Sometimes the movements become more complex with an individualized sequence of patting, rubbing, or twirling. These stereotypies may last for hours. Though the cause of the repetitive movements is unclear, they seem to have a calming effect and may (especially in the older child) surface in times of stress. This repetitiveness is reflected in a rigid need for sameness in daily routines. Children with autism can develop elaborate rituals in which the order of events, the exact words, and the arrangement of objects must be followed. Failure of parents/caretakers to follow the proscribed order of events results in inconsolable outbursts.

Other symptoms occurring in a substantial number of individuals with autism spectrum disorders:

*

Hyper- and hyposensitivities to sound and touch. Loud or high-pitched noises such as the vacuum cleaner cause great discomfort, causing the child to hold his hands over his ears. The feel of certain clothes or of being touched may be unbearable; conversely, truly painful stimuli like a burn or laceration are ignored.
*

Odd behaviors around foods and their presentation, such as accepting a limited number of foods or only eating french fries that come from McDonalds®.
*

Abnormal sleep patterns (60%), such as never sleeping through the night, trouble going to sleep, or getting up for the day at 2:00 am.
*

Tantrums and/or self-injurious and aggressive behaviors brought on by a change in routine, an offending touch, being asked to do something they do not want to do, or for no apparent reason.
*

Impaired motor development with toe walking early in life, hypotonia, general clumsiness, and inability to ride a two-wheel bicycle.
*

Total disregard for danger, resulting in high risk of early death, commonly from drowning.

Evidence suggests that as many as 15% of adolescents or young adults with autism develop a catatonia syndrome associated with marked deterioration in movement with slowness and freezing in mid-movement, vocalizations, and regression of self-care skills.

Obesity is a common complication of unknow cause. Medication side-effects, inactive life style and difficulty withholding food from children with aggression may be implicated.

Delineation of ASD subgroups. In an attempt to sort out the clinical and etiologic heterogeneity within autism, researchers are increasingly emphasizing the identification of phenotypic features (endophenotypes or biomarkers) to delineate subgroups that could predict outcomes and direct treatment choices [Viding & Blakemore 2007]. The terms “endophenotypes” and “biomarkers” imply that these neurophysiologic, biochemical, endocrinologic, neuroanatomic, and cognitive features are more biologic and, thus, more proximally related to the underlying etiologic processes than the behavioral symptoms [Gottesman & Gould 2003].

The following phenotypes have been investigated:

*

IQ scores in longitudinal studies strongly predict long-term outcomes and are directly associated with the degree of autistic psychopathology even in young children [Lord et al 2001, Howlin et al 2004, Chawarska & Bearss 2008]. Stevens et al [2000] reported that early normal or near-normal nonverbal IQ is the best predictor of adequate functioning by grade school; however, in the presence of significant language addition, uneven cognitive profiles typical of autism may make an average of widely discrepant scores meaningless [Klin et al 2005a]. That said, 50% to 70% of autistic children have historically been classified as intellectually disabled by nonverbal IQ testing [Fombonne 2005, Baird et al 2006]. An epidemiologic study from the Centers for Disease Control and Prevention reported 44.6% of the ASD population had intellectual disability. Lower rates of intellectual disability reported in more recent studies are probably the result of broadening of the ASD diagnostic criteria to include Asperger syndrome and children with milder symptoms [Autism and Developmental Disabilities Monitoring Network 2009].
*

Seizures develop in approximately 25% of children with autism. In addition, the rate of electroencephalographic abnormalities is increased in children with autism who do not have a history of seizures [Kim et al 2006, Spence & Schneider 2009]. As in the general population, seizures alone are not a sensitive predictor of outcome. However, the prevalence of seizures is higher among individuals with moderate to severe intellectual disability and those with motor deficits [Tuchman & Rapin 2002]. And individuals with autism plus epilepsy have on the average lower IQs and poorer adaptive, behavioral, and social outcomes than those without epilepsy [Hara 2007].
*

Structural brain malformations, typically identified by MRI, usually portend a poorer outcome [Miles & Hillman 2000]. In a recent study of 77 children with autistic disorder of unknown cause and uncomplicated by seizures, severe intellectual disability, major anomalies, or focal neurologic signs, neuroradiologists reported that 40% had some abnormality, of which white-matter signal abnormalities, severely dilated Virchow-Robin spaces, and temporal lobe structural abnormalities were the most common [Boddaert et al 2009]. This level of pathology in children with nonsyndromic autism lends support to the controversial recommendation to obtain brain MRIs as a standard diagnostic test in autism.
*

Significant generalized dysmorphology, found in 15% to 20% of individuals with autism, is a reliable indicator of an insult to early development [Miles & Hillman 2000]. Using a classification system that defines generalized dysmorphology, Miles et al [2005] found that in 81% of individuals dysmorphology was predictive of a poor outcome (defined as nonverbal with IQ <55). The presence of significant dysmorphology was also the top predictor of a poor response to early intensive behavioral therapy [Stoelb et al 2004]. The Autism Dysmorphology Measure (ADM), which guides clinicians through an assessment of 12 body areas, was developed to provide non-geneticists with a standardized method for assessing generalized dysmorphology [Miles et al 2008]. The 12 areas assessed are: height, hair growth pattern, ear structure, size and placement, nose size, face size and structure, philtrum, mouth and lips, teeth, hand size, fingers and thumbs, nails, and feet. * Microcephaly (head circumference <2nd centile) occurs in 5%-15% of children with autism [Fombonne et al 1999, Miles et al 2000, Miles et al 2005] and is highly predictive of poor outcome. * Macrocephaly (head circumference greater than the 97th centile), found in approximately 30% of children with autism, does not strongly correlate with outcome or IQ [Miles et al 2000] though Lainhart noted an association with delay in acquisition of first words [Lainhart et al 2006]. Using dysmorphology and microcephaly, autism can be defined as complex or essential [Miles et al 2005]: * Complex autism is defined by the presence of dysmorphic features and/or microcephaly, features which indicate some alteration of early morphogenesis. Approximately 20%-30% of children ascertained because of a diagnosis of autism have complex autism. Complex autism is associated with a poorer prognosis, a lower male-to-female ratio, and a lower sibling recurrence risk than essential autism. Approximately 30% of children with complex autism can be diagnosed with an autism associated syndrome or chromosome disorder using currently available diagnostic tests (see Causes of Autism) [Miles et al 2008]. * Essential autism is defined by the absence of generalized dysmorphology and microcephaly. Approximately 70%-80% of children with autism have essential autism. Children with essential autism are more likely to be male, to have a higher sibling recurrence risk, and to have a greater family history of autism and autism-related disorders such as alcoholism and depression than children with complex autism. As a group, the outcome is better for essential autism than complex autism, though most children with essential autism still do poorly. Currently available testing is less likely to reveal an exact etiologic diagnosis in essential autism than in complex autism. Establishing the Diagnosis The American Psychiatric Association Manual of Psychiatric Diseases, 4th edition, is the primary diagnostic reference for autism used in the US. The update in 2000 changed some of the accompanying text but did not change the diagnostic criteria. (See American Psychiatric Association [2000].) The DSM-IV in 1994 placed autistic disorder, Asperger syndrome, Rett syndrome, childhood disintegrative disorder, and pervasive developmental disorder-not otherwise specified (PDD-NOS) under the umbrella diagnostic term ‘pervasive developmental disorders.’ Subsequently, discovery of MECP2 mutations as the cause of Rett syndrome, uncertainty about nosology of childhood disintegrative disorder, and better understanding of the continuity within the autism diagnoses led to adoption of the term autism spectrum disorders (ASD) as the favored umbrella designation for autistic disorder (AD), Asperger syndrome (AS) and PDD-NOS (National Institute of Mental Health). It is expected that the DSM-V, due out in 2012, will further simplify the nosology. Autistic Disorder— DSM-IV Diagnostic Criteria (diagnostic code 299.00) I. A total of six (or more) items from A, B, and C, with at least two from A, and one each from B and C: A. Qualitative impairment in social interaction, as manifested by at least two of the following: B. Qualitative impairments in communication as manifested by at least one of the following: 1. Marked impairment in the use of multiple nonverbal behaviors such as eye-to-eye gaze, facial expression, body postures, and gestures to regulate social interaction 2. Failure to develop peer relationships appropriate to developmental level 3. A lack of spontaneous seeking to share enjoyment, interests, or achievements with other people (e.g., by a lack of showing, bringing, or pointing out objects of interest) 4. Lack of social or emotional reciprocity 5. Delay in, or total lack of, the development of spoken language (not accompanied by an attempt to compensate through alternative modes of communication such as gesture or mime) 6. In individuals with adequate speech, marked impairment in the ability to initiate or sustain a conversation with others 7. Stereotyped and repetitive use of language or idiosyncratic language 8. Lack of varied, spontaneous make-believe play or social imitative play appropriate to developmental level 9. Encompassing preoccupation with one or more stereotyped and restricted patterns of interest that is abnormal either in intensity or focus 10. Apparently inflexible adherence to specific, nonfunctional routines or rituals 11. Stereotyped and repetitive motor mannerisms (e.g., hand or finger flapping or twisting or complex whole-body movements) 12. Persistent preoccupation with parts of objects C. Restricted repetitive and stereotyped patterns of behavior, interests, and activities, as manifested by at least one of the following: II. Delays or abnormal functioning in at least one of the following areas, with onset prior to age three years: 1) social interaction, 2) language as used in social communication, or 3) symbolic or imaginative play III. The disturbance is not better accounted for by Rett syndrome or childhood disintegrative disorder. Asperger syndrome [Asperger 1944, Frith 1991 (English translation)] is characterized by relatively normal language development (including timing, grammar, and vocabulary) but requires all other DSM-IV diagnostic criteria for autism. Individuals with Asperger syndrome are generally loners, are uncomfortable in groups, are unable to empathize with others, do not chat, follow a literal interpretation of speech with no understanding of idioms or jokes, maintain a sameness in routine, follow strict rules, and have an encompassing preoccupation with one domain, such as the weather or computers. Speech may be pedantic or repetitive with odd intonations. IQ must be within the normal range to qualify for the diagnosis. Clumsiness is common. Whether Asperger syndrome is the expression of the high end of the autism spectrum or a discrete genetic entity is unclear. The diagnosis of Asperger syndrome is problematic, with poor concordance between the various diagnostic instruments [Klin et al 2005b]. The Autism Spectrum Screening Questionnaire (ASSQ) [Ehlers et al 1999], the Asperger Syndrome Diagnostic Interview (ASDI) [Gillberg et al 2001], the Australian Scale for Asperger's Syndrome [Garnett & Atwood 1997], and the Childhood Asperger Syndrome Test (CAST) [Scott et al 2002] are also available. A recent video, Asperger's Diagnostic Assessment [Attwood 2004] provides a hands-on tutorial which should be useful to the clinician. Pervasive developmental disorder - not otherwise specified (PDD-NOS). Children with autistic symptoms who do not meet full criteria in all three diagnostic domains can be diagnosed with PDD-NOS. This includes children with milder symptoms of all three autism diagnostic criteria and those meeting full criteria for autism in two of the three domains. Sometimes PDD-NOS is used as an initial or tentative diagnosis for younger children or before diagnostic evaluations are completed. Broader autism phenotype (BAP) may designate siblings or other family members with some autism symptoms [Piven & Palmer 1999, Pickles et al 2000, Geschwind et al 2001]. This terminology was originally adopted by researchers to classify sibs who were likely to have mutations in putative autism genes and reflects the growing awareness of the broad phenotypic spectrum of autism spectrum disorders. Childhood disintegrative disorder is a rare condition manifesting before age ten years in which children who have developed normally for at least two years deteriorate and lose previously acquired language, social, and play skills. The condition may resemble autism in clinical presentation but differs from autism in the pattern of onset, course, and outcome. It is no longer considered one of the pervasive developmental disorders. Diagnostic and Screening Tools To diagnose autism, one must precisely enumerate the autism symptoms and their age of occurrence, which can be done using the DSM-IV or a number of checklists [Filipek et al 2000, Cavagnaro 2007]: * CARS (Childhood Autism Rating Scale) [Schopler et al 1986] consisting of 15 questions scored by the parent and the tester is the most commonly used diagnostic checklist. It is a reliable, well-verified measure which is relatively fast and easy to administer. A score of 30 to 35 indicates mild autism and 36 or higher moderate-to-severe autism. * ABC (Autism Behavior Checklist) [Aman et al 1985] * GARS (Gilliam Autism Rating Scale) [Gilliam 1995] In North America, research criteria for the diagnosis of autism depend primarily on the ADI-R (Autism Diagnostic Interview-Revised) [Lord et al 1994], which is a detailed parent interview, and the somewhat shorter ADOS (Autism Diagnostic Observation Schedule) [Lord et al 1989]. Both scales follow the DSM-IV criteria and were developed in an attempt to sort autism by its behavioral symptoms to permit identification of homogeneous populations. These scales are not widely used in clinical practice because of the time and expense to administer, although the shorter ADOS is becoming more widely used outside of research settings. To guide allocation of services school systems use educationally based “autism eligibility” criteria which are similar but not identical to the medical diagnostic criteria, sometimes leading to conflicts. This is particularly true for the higher-functioning or Asperger syndrome students whose behavioral manifestations need remediation through the schools, but may not meet the educational egibility criteria. It is recommended that all children be screened for autism by their primary health care provider. A positive score on one of these tools is not diagnostic for an ASD but prompts referral to a diagnostic clinic. * M-CHAT (Checklist for Autism in Toddlers-modified) [Robins et al 2001] is the most commonly used screening tool. This 23 item checklist, designed for primary care providers to identify at-risk toddlers at the 18-month visit, can be filled out by parents in the waiting room and is available in Spanish and English [Cavagnaro 2007]. A recent replication study [Kleinman et al 2008] confirmed the validity in detecting possible ASD in both low- and high-risk groups aged 16 to 30 months. It is recommended by the Neurology Quality Standards Subcommittee [Filipek et al 1999, Filipek et al 2000]. * Infant/Toddler Checklist (pdf) from the Communication and Symbolic Behavior Scales Developmental Profile [Wetherby & Prizant 2002] is recommended by the American Academy of Pediatrics to identify at-risk children younger than age 18 months [Johnson & Myers 2007]. Prevalence An increase in the prevalence of all the autism spectrum disorders is being reported worldwide. * Prior to 1990, most studies estimated a general population prevalence for autism of four to five per 10,000 (1/2000-1/2500) [Fombonne 2001]. * During the 1990s, studies of preschool children in Japan, England, and Sweden reported prevalence rates for autism of 21 to 31 per 10,000 (1/476 -1/323) [Arvidsson et al 1997, Baird et al 2000]. * A CDC case-finding study in Brick Township, New Jersey reported prevalence at 40 per 10,000 (1/250) for autism and 67 per 10,000 (1/149) for all PDDs [Autism and Developmental Disabilities Monitoring Network 2009]. * An epidemiologic study from the United Kingdom utilizing specialized visiting nurses who monitor child health and development at ages seven months, 18 to 24 months, and three years reported a prevalence rate of 16.8 per 10,000 (1/595) for autism and 63 per 10,000 (1/159) for all PDDs in children younger than age five years [Chakrabarti & Fombonne 2001]. Those rates were confirmed, reporting a prevalence rate of 22 per 10,000 (1/455) for autism and 59 per 10,000 (1/169) for all PDDs in children younger than age six years [Chakrabarti & Fombonne 2005]. * Two recent studies in the United States reported the diagnosis of an ASD in 1/91 children age three to seventeen years [Kogan et al 2009] and 1/110 children age eight years [Autism and Developmental Disabilities Monitoring Network 2009]. Recent analyses indicate that the “autism epidemic” does not reflect a true increase in the incidence of ASD, but is attributable to increased awareness by both the public and professionals, leading to more complete case finding together with broadening of the diagnostic criteria [Gernsbacher et al 2005, Fombonne et al 2006, Shattuck 2006, Taylor 2006, Atladottir et al 2007]. Studies finding the greatest increase in ASD also note lower rates of intellectual disability in these children. Only 30% of children with PDDs ascertained by Chakrabarti & Fombonne [2005] were intellectually disabled compared with 70% of children in earlier studies. This suggests that many of the higher-functioning children with milder autistic symptoms, such as less severe language impairment and fewer aggressive behaviors had not been counted in past epidemiologic surveys. A recent update on the ongoing epidemiologic surveillance of autism in California indicated that the rise of autism in California shows no sign of plateauing. The study concluded that earlier age at diagnosis and inclusion of milder cases accounted for more than two-thirds of the increase but stated that the extent to which the continued rise could represent a true increase in the occurrence of autism remains unclear [Hertz-Picciotto & Delwiche 2009]. Causes of Autism

Twin and family studies have established the preponderant genetic basis of autism and indicate that the heritability of autism is over 90% [Monaco & Bailey 2001]. Currently a genetic cause can be identified in 20% to 25% of children with autism. A small number of cases of autism can be traced to specific teratogenic exposures. The cause of autism in the remaining 75% to 80% remains unknown.

The growing number of distinct, individually rare genetic causes of autism suggests that the genetic basis of autism resembles that of intellectual disability and cerebral palsy with many syndromes, each individually rare, implicated in the development of autism. But it also seems likely that complex and as-yet-undetermined interactions between “small-effect” inherited changes will also be found to be causative.
Genetic Causes

Known genetic causes of autism include:

*

Cytogenetically visible chromosomal abnormalities (~5%)
*

Copy number variants (CNVs) (i.e., submicroscopic deletions and duplications) (10%-20%)
*

Single gene disorders in which neurologic findings are associated with ASD (~5%)

Cytogenetically Visible Chromosomal Abnormalities

High-resolution chromosome analysis reveals chromosome aneuploidy in approximately 5% of children with ASD. Another 3%-5% have identifiable chromosomal abnormalities using FISH techniques. As expected, unbalanced chromosome abnormalities are found predominantly in children with autism and accompanying dysmorphology [Miles et al 2005, Jacquemont et al 2006, Takahashi & Miles 2009].

Although cytogenetic abnormalities on almost every chromosome have been found in autism, only a few occur commonly enough to be possible loci for autism genes [Wassink et al 2001, Reddy 2005, Vorstman et al 2006]. A curated database of chromosome abnormalities reported in individuals with autism is available at projects.tcag.ca/autism [Marshall et al 2008].

Maternally derived duplication of the Prader-Willi/Angelman syndrome critical region (15q11-q13) is the most commonly observed chromosome abnormality in autism, detected in 1%-3% of children with autism. Most commonly this duplication is the result of a de novo supernumerary isodicentric 15q chromosome and less commonly the result of segregation of a parental chromosome translocation or a maternally derived interstitial 15q duplication. Routine cytogenetic analysis detects supernumerary isodicentric 15q but the diagnosis of interstitial duplications requires interphase FISH analysis or aCGH. The maternally derived 15q11-q13 interstitial duplication is a highly penetrant cause of autism, whereas the paternally derived duplication has little or no phenotypic effect, indicating the significance of genomic imprinting of this region [Hogart et al 2010].

A maternal duplication of 15q, resulting in trisomy for that region, causes subtle effects on the physical phenotype whereas,children with four copies of 15q including those with a supernumerary isodicentric 15 are typically more impaired and may exhibit hypotonia, seizures, microcephaly, and severe developmental delay [Borgatti et al 2001, Dykens et al 2004, Hogart et al 2010].

Trisomy 21. Children with Down syndrome have autism more commonly than expected. The incidence was at least 7% in one study [Kent et al 1999].

45, X Turner syndrome. Girls and women with Turner syndrome who have a maternal X chromosome (45,Xmat) have poorer social cognition skills than girls who have a paternal X chromosome (45,Xpat) [Skuse 2000].

Other. Chromosome abnormalities reported more than once include deletions of 2q37, 18q, 22q13.3, Xp22.3, and the sex chromosome aneuploidies 47,XYY, 47,XXY, and 45,X [Gillberg 1998, Manning et al 2004, Vorstman et al 2006, Jha et al 2007, Marshall et al 2008, Shinawi et al 2009a]. All reported terminal deletions of 2q37 and 22q13.3 have been associated with dysmorphic phenotypes [Lukusa et al 2004, Manning et al 2004]. Characterization of the minimal critical chromosomal regions for the 22q13.3 and the Xp22.3 deletions in persons with autism of unknown cause has led to the identification of mutations in the SHANK3 [Durand et al 2007] and NLGNX4 [Jamain et al 2003] genes, respectively.
Copy Number Variants (CNVs)

Array comparative genomic hybridization (aCGH) is steadily replacing high-resolution chromosome analysis and FISH in the evaluation of children with autism. Clinically available platforms used in aCGH are designed to test for known deletion/duplication syndromes on the entire genome plus assessment of subtelomeric regions. The platforms used in aCGH vary in the density and type of molecular markers (BAC, SNP, and oligonucleotide clones) and are constantly being upgraded as new CNV hot spots are identified.

Currently, aCGH identifies clinically relevant de novo genomic imbalances in 7%-10% of individuals with autism of unknown cause [Sebat et al 2007, Christian et al 2008, Kumar et al 2008, Marshall et al 2008, Weiss et al 2008]; the yield was higher in those whom the authors identified as having “syndromic” autism.

*

Using a 1-Mb genome-wide array, Jacquemont et al [2006] identified clinically relevant CNVs in 27.5% (8/29) of individuals with autism and dysmorphology, who previously had a normal karyotype as determined by routine cytogenetic studies.
*

Using an oligonucleotide array, Sebat et al [2007] found de novo copy number changes in 10% of children from simplex families (i.e., autism in a single family member) and 2% from multiplex families (i.e., autism in multiple family members) compared to 1% in controls.
*

Using a dense genome-wide SNP array, Marshall et al [2008] found unbalanced CNVs in 44% of 427 unrelated families with autism that were not present in control families. Many of these CNVs were inherited and only 7% were de novo in persons with autism of unknown cause.
*

A whole-genome CNV study using 550,000 SNP markers on a cohort of 859 individuals with ASD and 1,409 healthy children of European ancestry revealed several pathogenic genomic changes in genes encoding neuronal cell-adhesion molecules (NRXN1, CNTN4, NLGN1, and ASTN2) and in genes involved in the ubiquitin pathways (UBE3A, PARK2, RFWD2, and FBXO40) [Glessner et al 2009].

These studies highlight the potential of array-based techniques. At this time, however, caution must be exercised in interpreting these results and their relation to autism. The de novo occurrence of a copy number variation is not absolute evidence of its pathogenicity. Distinguishing characteristics of pathogenic CNVs include (1) de novo event not found in either parent or inherited from an affected parent, (2) deletions or duplications which include genes known to be expressed in the brain, (3) large CNVs, and (4) deletions, as opposed to duplications

Some CNVs associated with autism include the following:

*

16p11.2 deletion syndrome is characterized by developmental delay, intellectual disability, and/or autism spectrum disorder (ASD). Developmental delays are more related to diminished language and cognitive function than motor disability. Although IQ scores range from mild intellectual disability to normal, those with IQ scores in the average range typically have other developmental issues such as language delay or ASD. Expressive language appears to be more affected than receptive language. Weiss et al [2008] reported 16p11.2 deletions or duplications in approximately 1% of individuals with autism and 1.5% of children with developmental or language delays. Marshall et al [2008] observed a similar 1% frequency.

The 16p11.2 deletion often occurs de novo, but may be transmitted from parent to child in an autosomal dominant manner. Of note, however: the same 16p11.2 CNVs can be observed in a variety of other disorders including schizophrenia, bipolar disorder, seizures, ADHD, and dyslexia, as well as apparently unaffected family members; thus, interpretation of the significance of this CNV can be difficult [McCarthy et al 2009, Rosenfeld et al 2010, Shinawi et al 2010].

This CNV is located at a hot spot of genomic instability caused by duplicated blocks of DNA that lead to unequal crossing over during meiosis and is detectable by clinical oligonucleotide aCGH platforms and some bacterial artificial chromosome (BAC)-based platforms. Other test methods that can detect this deletion include multiplex ligation-dependent probe amplification (MLPA), metaphase fluorescence in situ hybridization (FISH), and quantitative polymerase chain reaction PCR (qPCR).
*

15q13.3 deletion syndrome has been associated with intellectual disability and epilepsy [Sharp et al 2008] and ASD [Ben Shachar et al 2009, Miller et al 2009, Pagnamenta et al 2009] and appears to be clinically variable. More recent data suggest that haploinsufficiency of CHRNA7 is causative for the majority of neurodevelopmental phenotypes in the 15q13.3 microdeletion syndrome [Shinawi et al 2009b].

Single Gene Disorders

The following singe gene disorders are commonly observed to cause syndromic autism.

Fragile X syndrome. Whereas 1% to 3% of children ascertained on the basis of an autism diagnosis have fragile X syndrome, at least half the children with fragile X syndrome have some autistic behaviors, including avoidance of eye contact, language delays, repetitive behaviors, sleep disturbances, tantrums, self-injurious behaviors, hyperactivity, impulsiveness, inattention, and sound sensitivities. In one study of 63 males with fragile X syndrome, 30% met criteria for autistic disorder and 30% criteria for PDD-NOS [Harris et al 2008].

Fragile X syndrome is caused by expansion of the CGG trinucleotide repeat in the FMR1 gene to the full mutation size of 200 or more CGG repeats.

A considerable number of children being evaluated for autism are found to have FMR1 premutations (55-200 CGG repeats) [Goodlin-Jones et al 2004, Cornish et al 2005, Reddy 2005, Farzin et al 2006, Loesch et al 2007]. Farzin et al [2006] studied 14 boys with premutations ascertained through an autism clinic and found 71% met ASD diagnostic criteria. In the authors’ experience, ten of 488 (2%) persons ascertained through a dedicated autism clinic had either an FMR1 full mutation or premutation. Of the five children with a full mutation, only one was diagnosed with autistic disorder; four did not meet criteria for an ASD diagnosis. For the five premutation carriers, four were diagnosed with autistic disorder and one with PDD-NOS [Takahashi & Miles 2009]. This underscores the importance of performing FMR1 molecular genetic testing in all children being evaluated for an autism spectrum disorder.

Molecular studies indicate the FMR1 gene may cause the autism phenotype via two mechanisms: RNA toxicity to the neurons and gene silencing that affects neuronal connectivity [Schenck et al 2003, Handa et al 2005, Hagerman et al 2008].

PTEN macrocephaly syndrome. The PTEN (phosphatase and tensin homolog) gene was initially described as a tumor suppressor gene associated with a broad group of disorders referred to as PTEN hamartoma tumor syndrome which includes Cowden syndrome, Bannayan-Riley-Ruvalcaba syndrome, Proteus syndrome, and Lhermitte-Duclos disease. More recently, PTEN gene mutations have been associated with autism and macrocephaly [Zori et al 1998, Parisi et al 2001, Delatycki et al 2003, Butler et al 2005, Buxbaum et al 2007a]. Correspondingly, PTEN is recognized to play an important role in brain development, including neuronal survival and synaptic plasticity.

The frequency of PTEN mutations as a cause of ASD is unclear; results from studies of children ascertained through autism and macrocephaly range from 1% [Buxbaum et al 2007a] to 8.3% [Varga et al 2009] to 17% [Butler et al 2005]. Both de novo and familial PTEN mutations have been identified in this population. It may be significant that more of the children studied by Buxbaum et al [2007a] were from multiplex families, whereas the children studied by Butler et al [2005] were from simplex families.

Children with ASD found to have a PTEN mutation generally have extreme macrocephaly ranging from +3.7 SD to +9.6 SD (average: +5.4 SD) [Buxbaum et al 2007a]. In addition, mutation of PTEN is not specific for autism: Varga et al [2009] found children with macrocephaly and intellectual disability but not autism had a similar chance of having a PTEN mutation.

Because PTEN germline mutations are associated with the phenotypically broad PTEN hamartoma tumor syndrome, it is recommended that children with PTEN mutations and their families be evaluated by a medical geneticist for clinical signs of any of the related disorders. Moreover, because these disorders carry a risk of cancer, including cancer of the breast, thyroid, endometrium, and kidney, these individuals need to be involved in a tumor surveillance program. See PTEN Hamartoma Tumor Syndrome.

Sotos syndrome. Sotos syndrome is characterized by the cardinal features of typical facial appearance, overgrowth (height and head circumference ≥2 SD above the mean), and learning disability ranging from mild (children attend mainstream schools and are likely to be independent as adults) to severe (lifelong care and support are required). Sotos syndrome is associated with the major features of behavioral problems, congenital cardiac anomalies, neonatal jaundice, renal anomalies, scoliosis, and seizures. Though Sotos syndrome is probably not a significant cause of classic autism [Buxbaum et al 2007b], children with Sotos syndrome and behavioral problems such as difficulty with peer group relationships and lack of awareness of social cues may be referred to autism clinics.

Eighty to 90% of individuals with Sotos have a demonstrable mutation or deletion of NSD1; inheritance is autosomal dominant.

Rett syndrome is one of the original DSM-IV-designated pervasive developmental disorders and the only one for which a specific genetic etiology has been identified [Amir et al 1999]. Ninety-six percent of individuals with classic Rett syndrome have mutations in the X-linked MECP2 gene [Moretti & Zoghbi 2006]. Though the vast majority of individuals with MECP2 mutations are girls, MECP2 mutations are identified in 1%-2% of males with developmental disorders, including infantile encephalopathy, intellectual disability with motor deficits and early-onset bipolar disorder and schizophrenia. Most males are identified in families with an X-linked pattern of intellectual disability.

The phenotype of MECP2-confirmed Rett syndrome overlaps considerably with autism of unknown cause; children with both often have a period of normal development followed by loss of language with stereotypic hand movements. However, Rett syndrome can usually be distinguished clinically based on a decreasing rate of head growth, progressive gait disturbance, and hand-wringing in early childhood. Initially, the distinction may be difficult: a study of two Rett syndrome databases found that 17.6% (55/313) of girls with MECP2-confirmed Rett syndrome had been given an early diagnosis of autism [Young et al 2008]. Those girls had significantly milder Rett syndrome symptoms, were more likely to remain ambulatory, retained some functional hand use, and developed specific Rett syndrome symptoms later. They were also more likely to have the mutation p.Arg306Cys or p.Thr158Met. The recommendation of Young et al [2008] that all girls diagnosed with autism be monitored carefully for evolving signs of Rett syndrome (including a deceleration in head growth) seems justified.

Overall, MECP2 mutations have been reported in approximately 1% of children diagnosed with autism [Moretti & Zoghbi 2006, Lintas & Persico 2009]. It is not clear at this time whether this justifies MECP2 molecular genetic testing in girls diagnosed with apparently classic autism, especially girls who will be followed on a regular basis in a clinic with expertise in both disorders.

Evidence of variable expression of the protein MeCP2 in the brains of individuals with both autism and Rett syndrome and evidence that MeCP2 deficiency can reduce expression of the genes UBE3A and GABRB3 implicated in autism, indicate some causal relationship between the two disorders [Samaco et al 2004, Samaco et al 2005]. Intensive study of the role of MeCP2 in maintaining neuronal function and evidence of symptom reversal of neuronal symptoms in mice following reactivation of the silenced Mecp2 gene is projected to have implications for autism treatment [Bird 2008].

Tuberous sclerosis complex (TSC). Although 25%-50% of intellectually disabled individuals with TSC fulfill autism diagnostic criteria, only 1.1%-1.3% of individuals initially diagnosed with ASD have TSC [Fombonne et al 1997b, Baker et al 1998, Asano et al 2001, de Vries et al 2007]. Early-onset infantile spasms and temporal lobe tubers on MRI examination increase the chance that children with TSC2 mutations will also develop autism [Bolton 2004].

In a prospective study of children with TSC evaluated using the ADOS, 66% of infants met criteria for autism or ASD at age 18 months, 54% at age 24 months, 46% at age 36 months, and 50% at age 60 months.The children with both TSC and autism were more cognitively impaired than those with TSC only [Jeste et al 2008].

An evaluation for signs of TSC including skin lesions (hypopigmented macules, shagreen patches, adenoma sebaceum) and a family history consistent with autosomal dominant inheritance of findings suggestive of TSC (seizures, skin lesions, intellectual disability) are generally sufficient to indicate or rule out the diagnosis of TSC in children with autism. Molecular genetic testing for the two causative genes, TSC1 and TSC2, is clinically available. Recurrence risks for families with a child with autism associated with TSC may be significantly higher than for for families with autism of unknown cause. Mechanisms underlying autism in TSC are unknown.

Neurofibromatosis type 1 (NF1). Although NF1 has been diagnosed in children with autism, it is unclear whether this is a true association or the chance simultaneous occurrence of two relatively common childhood disorders [Fombonne et al 1997b, Battaglia & Carey 2006, Schaefer & Lutz 2006]. No genetic overlap between mutations in the NF1 gene and autism has been demonstrated [Zafeiriou et al 2007].

Timothy syndrome. Timothy syndrome, a disorder of calcium channels caused by a mutation in the CACNA1C gene, is characterized by severe QT prolongation, syndactyly, cardiac defects, dysmorphic faces, developmental delays, and autistic symptoms [Splawski et al 2004]. Inheritance is autosomal dominant.

Joubert syndrome. This autosomal recessive disorder is characterized by partial or complete agenesis of the cerebellar vermis, seen as the “molar tooth sign” on MRI, abnormal breathing, abnormal eye movement, cognitive impairment, and behavioral problems. A subset of Joubert syndrome appears related to the AHI1 gene, encoding the ‘jouberin’ protein [Alvarez Retuerto et al 2008]. In one study, three of 11 children with Joubert syndrome met diagnostic criteria for autism and one of 11 for PDD-NOS [Ozonoff et al 1999]. However, Takahashi et al [2005] delineated behavioral and genetic differences between autism and Joubert syndrome, implying that they are etiologically distinct disorders. In a report by Muhle et al [2004] of monozygotic twins with Joubert syndrome, the twin with the more severe cerebellar abnormality had autism, suggesting that some disorders may have the potential to cause the autism phenotype if as-yet unidentified autism regions or circuits of the brain are affected.

Metabolic conditions

*

Mitochondrial disorders. Although mitochondrial respiratory chain disorders have only been reported in individuals with autism on rare occasions, elevated plasma concentrations of lactate have been frequently noted [Coleman 2005, Correia et al 2006]. A population-based study of 69 children with autism reported an elevated plasma lactate concentration in 20% (14/69); five of the eleven children undergoing muscle biopsy had a deficiency of one or more respiratory chain complexes, most frequently complexes I, IV, and V, based on enzymatic complex activity less than 20% of normal [Oliveira et al 2005]. If confirmed, this would be the largest etiologic autism subgroup. Identifying a mitochondrial disorder is more likely in autistic children with atypical features such as hypotonia, failure to thrive, and intermittent episodes of regression than in children without these findings. Weissman et al [2008] analyzed data from 25 persons with a mitochondrial disorder and an initial diagnosis of autism and found they could all be distinguished from autism of unknown cause on the basis of an abnormal neurologic examination and/or an elevated plasma lactate concentration. In addition, mitochondrial dysfunction has also been reported in persons with ASD without additional neurologic features [Pons et al 2004, Smith et al 2009].
*

Phenylketonuria (PKU). Co-morbidity of ASD and untreated PKU has been described but the autism diagnosis is usually complicated by the severe intellectual disability found in these children. A systematic study showed that none of 62 persons with PKU who were diagnosed and treated early met diagnostic criteria for autism, whereas two of 35 (5.7%) persons with PKU who were diagnosed late fulfilled the diagnostic criteria for ASD [Baieli et al 2003].
*

Adenylosuccinate lyase deficiency. This rare autosomal disorder of de novo purine synthesis results in the accumulation of succinylpurines in body fluids. In about half of affected individuals the variable clinical manifestations include developmental delay, seizures, and autism symptoms including failure to make eye contact, repetitive behavior, agitation, temper tantrums, and aggression [Van den Berghe et al 1997]. In one study, one out of 420 children with PDD was found to have adenylosuccinate lyase deficiency [Stathis et al 2000].
*

Creatine deficiency syndromes (CCDSs). The CCDSs, inborn errors of creatine metabolism, include the two creatine biosynthetic disorders, guanidinoacetate methyltransferase (GAMT) deficiency and L-arginine:glycine amidinotransferase (AGAT) deficiency (or GATM deficiency), and the creatine transporter defect, SLC6A8 deficiency. Intellectual disability and seizures are common to all three CCDSs. Approximately 80% of individuals with GAMT deficiency have a behavior disorder that can include autistic behaviors and self-mutilation; approximately 45% have pyramidal/ extrapyramidal findings. Onset is between ages three months and three years. Only five individuals with AGAT deficiency have been reported. The phenotype of SLC6A8 deficiency in affected males ranges from mild intellectual disability and speech delay to severe intellectual disability, seizures, and behavioral disorder. Onset is between ages two and 66 years. Approximately 50% of females heterozygous for SLC6A8 deficiency have learning and behavior problems.

The prevalence of creatine deficiency syndromes in those with ASD appears to be low. Sequencing of the SLC6A8 gene in 100 males with ASD did not detect deleterious mutations [Newmeyer et al 2007].
*

Smith-Lemli-Opitz syndrome (SLOS). This autosomal recessive multiple congenital anomaly and intellectual disability syndrome is caused by a deficiency of 7-dehydrocholesterol reductase, an essential enzyme in the biosynthesis of cholesterol. SLOS can be associated with autism and other behavioral characteristics such as repeated self-injury, sensory hyper-reactivity, temperature dysregulation, and sleep disturbance [Tierney et al 2001, Tierney et al 2006]. Rates of autistic behavior reported in individuals with SLOS range from 50% to 86% [Manzi et al 2008]. One study found that three-fourths of the children with SLOS had ASD, about 50% diagnosed with autistic disorder and the rest with PDD-NOS [Sikora et al 2006]; no correlation was found between the abnormal metabolites and the presence or severity of autistic symptoms.

Other single gene disorders. Autism or autistic features have been described in children with many other single gene disorders. Most are associated with severe intellectual disability and significant dysmorphology and rarely are referred for medical evaluation with an initial question of autism. The list includes:

*

Cohen syndrome [Howlin et al 2005]
*

Cole Hughes macrocephaly syndrome [Naqvi et al 2000]
*

San Filippo syndrome
*

Cornelia de Lange syndrome
*

Angelman syndrome [Sahoo et al 2006, Bonati et al 2007]
*

Williams syndrome [Klein-Tasman et al 2007] and its reciprocal 7q11.23 microduplication syndrome [Berg et al 2007, Van der Aa et al 2009]
*

17p11.2p11.2 duplication syndrome [Potocki et al 2007]
*

22q11.1 deletion syndrome [Niklasson et al 2009]
*

WAGR (wilms tumor, aniridia, genitourinary anomalies and mental retardation) syndrome [Xu et al 2008]
*

Duchenne muscular dystrophy [Wu et al 2005b]

Developmental Syndromes of Undetermined Etiology—Commonly Observed

Moebius syndrome or sequence. Defined by unilateral or bilateral palsy of the sixth and seventh cranial nerves, Moebius syndrome is characterized by facial paralysis with inability to smile and fully abduct the eyes.It is often associated with abnormal tearing, seizures, hearing loss, and limb anomalies. About 30% of children with Moebius syndrome develop ASD [Johansson et al 2001, Stromland et al 2002, Bandim et al 2003]. A recent study confirmed the observations of Johansson et al [2001] that ASD occurs more frequently in individuals with Moebius syndrome with concurrent intellectual disability [Briegel et al 2009]. Presumably caused by an early disruption of embryonic blood supply leading to brain stem disruption, Moebius syndrome has been compared to thalidomide embryopathy which also damages the sixth and seventh cranial nerve and causes autism.

Landau-Kleffner syndrome (LKS). A small subset of children with ASD and late regression have LKS. These children develop sudden or gradual isolated language regression associated with seizures (epileptic aphasia) and/or severe EEG abnormality in deep sleep [Cortesi et al 2007]. In general, both the seizures and language impairment improve with normalization of EEG abnormalities [Spence & Schneider 2009].
Environmental Causes

The search for environmental causes of autism has been driven by the considerable increase in autism prevalence recorded over the last 20 years and the incomplete concordance for autism in monozygotic (MZ) twins.

In utero exposures, including valproic acid, thalidomide, and misoprostol (an abortifactant commonly used in South America) are recognized causes of autism. The Liverpool and Manchester Neurodevelopment Group recently reported a long-term study of 632 children exposed to antiepileptic drugs (AEDs) during gestation and found that children exposed to valproate in utero were seven times more likely to develop autism than those not exposed to AEDs. None of the families had a known family history of autism. They recommend that women taking valproate be informed of the risk for autism in children exposed during gestation [Bromley et al 2008].

Other factors that have been considered as causes of autism include expanded use of assisted reproductive technologies (ART) [Knoester et al 2007] and tocolytic drugs such terbutaline [Connors et al 2005].

Childhood immunizations given around the time that regressive-onset autism is recognized have been a focus of concern. Organic mercury, which constitutes roughly 50% of the preservative thimerosal used in certain injectable vaccines, and the measles-mumps-rubella (MMR) vaccine, which never contained mercury, have been studied. Though parental concern is still significant, multiple studies and lines of scientific evidence have identified no support for a relationship between immunizations and autism [DeStefano & Thompson 2004, Institute of Medicine 2004, Taylor 2006, Schechter & Grether 2008]. The original studies by Wakefield et al [1998] and Wakefield [1999] suggesting an associated between immunizations and autism have been disproved and the work was retracted by the journal The Lancet [Murch et al 2004]. One of the tragedies resulting from fear of an autism epidemic was the decreased use of childhood immunizations leading to out outbreaks of measles and childhood deaths [Jansen et al 2003, Offit 2008].
Multifactorial Inheritance

The heritability estimate of autistic disorder, calculated from recurrence risk data and monozygotic (MZ): dizygotic (DZ) twin concordance data, is more than 90%. Many have considered autism of unknown cause a multifactorial disorder based on the: (1) high heritability, (2) failure to identify major autism genes, (3) 4:1 male-to-female sex ratio, and (4) sibling recurrence risk of approximately 4% [Chakrabarti & Fombonne 2001, Gillberg et al 2001].

The multifactorial threshold model predicts that:

*

The more frequently affected sex (male) has a lower recurrence risk than the less frequently affected sex (female).
*

The less often affected sex (female) is more severely affected than the more often affected sex (male).

Studies indicate that autism does not follow this model:

*

Sibs of male and female probands with autism of unknown cause have the same risk of developing autism [Miles et al 2004].
*

The proportion of relatives with a mild subclinical autism phenotype, defined as increased impulsivity, aloofness, shyness, over-sensitivity, irritability, eccentricity and anxiety, is not increased when the proband is female [Pickles et al 2000].
*

When analyses are limited to probands with essential autism, females have less severe autistic symptoms than males [Miles et al 2004].

Lack of support for the classic multifactorial threshold model, however, does not eliminate the possibility that many genes are involved in autism. Instead, it suggests that genetic heterogeneity is the main impediment to understanding the inheritance of autism.
Genes that Cause or Increase the Risk of Autism

Determining specific genetic changes that increase the risk of developing autism is an area of intense study. The large-scale genetic studies of the last decade have ruled out the possibility that single gene of large affect causes a high proportion of autism. Rather it appears that a a significant number of highly penetrant autism alleles will be discovered in families. Study of these rare alleles will be essential to elucidating the pathogenetic mechanisms involved in the development of autism.

Array CGH has increased detection of copy number variants (CNVs) in autism and paved the way for identification of a number of new autism genes [Szatmari et al 2007]. Some CNVs may help identify highly penetrant causal mutations, while others may lead to the discovery of multiple common genetic variations which act in concert, possibly with environmental triggers to cause autism. Persuasive evidence for this common disease-common variant hypothesis in autism includes the presence of subtle sub-clinical autism symptoms in relatives of children with autism [Constantino et al 2009]. It is also probable that some CNVs may alter the expression of genes in the immediate vicinity of the CNV.

Table 1 which lists known and putative autism genes is unquestionably incomplete, as new candidate genes are being reported at an unprecedented rate. The genes are organized by pathogenesis to highlight the progress made in the functional assessment of autism candidate genes and pathways. In addition, some genes are included because of their compelling initial descriptionsthat still await confirmation. Selected references address both molecular and pathophysiologic descriptions.

Authoritative reviews of the current status of candidate genes and loci include: Veenstra-VanderWeele & Cook [2004], Wassink et al [2004], Grice & Buxbaum [2006], Gupta & State [2007], Szatmari et al [2007], Morrow et al [2008], O’Roak & State [2008], Sutcliffe [2008], Simons Foundation Autism Research Initeative [2009].

SFARI gene is a new comprehensive, Web-based, searchable list of candidate genes associated with ASD. The candidate genes are richly annotated for their relevance to autism, along with an in-depth, up-to-date view of their molecular function extracted from the current scientific literature.

The Genetic Association Database provides online access to human genetic association studies performed on autism and other complex disorders.

Evaluation Stretegy

In addition to behavioral assessment to establish the diagnosis of autism and cognitive testing, the evaluation strategy for all individuals with autism includes a medical evaluation to identify medical issues that affect the development and behavior of nonverbal children and a clinical genetics evaluation to elucidate diagnostic possibilities. This basic evaluation should be expanded if the medical or family history and/or physical examination raise concerns about metabolic, medical, or neurologic conditions. Recent reports from the American Academy of Pediatrics [Johnson & Myers 2007] and the American College of Medical Genetics [Schaefer & Mendelsohn 2008] provide practical approaches to the evaluation of children with ASD.

Family history. A three-generation pedigree should be obtained with attention to behavioral and neurologic diagnoses. Relatives with behaviors that are possible manifestations of autism may be examined directly or their records reviewed. Language, social, and psychiatric disorders, including alcoholism and possibly other addictive disorders, occur more often in relatives, including sibs, of clearly autistic probands, particularly those with essential autism [Fombonne et al 1997a, Bolton et al 1998, Piven & Palmer 1999, Pickles et al 2000, Miles et al 2003].

Clinical examination. Physical examination should include the following:

*

Measurement of height, weight, and occipital-frontal circumference (OFC) to identify (1) microcephaly or growth retardation which suggest various chromosome and monogenic syndromes, and (2) macrocephaly, which is present in approximately 35% of children with autism. In children with macrocephaly the molecularly defined macrocephaly syndromes that also cause autism, especially fragile X syndrome and PTEN hamartoma tumor syndrome, should be considered.
*

Dysmorphology examination to look for developmental anomalies which date the onset of the disorder. An in depth dysmorphology examination by a medical geneticist provides the best data on which to base the diagnostic plan. In addition, the brief Autism Dysmorphology Measure has been developed for use by non-dysmorphologists who evaluate children with autism [Miles et al 2008].
*

Skin examination (including Woods lamp examination) for evidence of tuberous sclerosis complex or neurofibromatosis type 1.
*

Assessment for signs of specific disorders known to be associated with autism:
o

PTEN mutation—macrocephaly, hamartomas or lipomas, freckles on the penis
o

Moebius syndrome—facial muscle weakness, incomplete abduction of eyes
o

Tuberous sclerosis complex—hypopigmented macules, shagreen patches, facial adenoma sebaceum
o

Fragile X syndrome—macroorchidism, long face, large ears
o

Mitochondrial disorders—failure to thrive, fatigue, hypotonia, recurrent episodes of regression

Laboratory testing. The following testing is recommended at the time of initial evaluation for all children with an ASD:

*

Array comparative genomic hybridization (aCGH) has replaced high-resolution chromosome analysis as the test of choice for the evaluation of any child with an autism spectrum disorder. Note: Cytogenetic analysis, in addition to aCGH, is recommended when the family history suggests transmission of a balanced chromosome rearrangement.
*

FMR1 molecular genetic testing for full mutations and premutations

The following should be considered based on the physical examination, review of systems, and family history:

*

Electroencephalogram if clinical signs of seizures or developmental regression are present. The utility of obtaining EEGs routinely is debated. It is clear that a significant number of children have EEG abnormalities and the likelihood of observing an abnormality increases with the duration of the EEG.
*

MRI. The brain MRI is indicated when the history and physical examination or neurologic examination suggests a localized lesion, tuberous sclerosis complex, Joubert syndrome, or an early environmental insult. Its routine use is controversial because of the expense and need for sedation or anesthesia by an anesthesiologist.
*

Metabolic testing. Metabolic evaluation, including quantitative plasma amino acids; urine organic acids; purines, creatine and guanidinoacetate in urine; serum concentration of lactate, pyruvate, creatine kinase, and uric acid; and CBC is of limited benefit for the majority of individuals with autism. However, because diagnosing and treating a metabolic disorder can significantly alter prognosis, selective and targeted metabolic work-up is recommended based on history and physical examination. The evaluation should include review of the child’s newborn screening test results. Further studies are needed to determine the prevalence of mitochondrial respiratory chain disorders in autism, especially those with recurrent setbacks, hypotonia, and failure to thrive.
*

Molecular genetic testing for a number of genes is clinically available and should be obtained if the phenotype and/or family history suggest the diagnosis

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.
Mode of Inheritance

Genetic counseling for families of probands with a chromosomal disorder, copy number variant, or a single gene disorder is based on information relevant to the primary diagnosis.

The mode of inheritance for autism of unknown cause is not known.
Risk to Family Members—Autism of Unknown Cause

Parents of a proband. No data on the risk to parents of a proband of having autism of unknown cause are available; however, parents of children with an ASD are more likely to exhibit mild autistic phenotypes, such as social awkwardness and a variety of psychiatric disorders including alcoholism, depression, obsessive-compulsive disorder, and panic and anxiety disorders when compared to parents of children with non-ASD disorders such as Down syndrome [Piven & Palmer 1999, Miles et al 2003, Wilcox et al 2003, Lauritsen et al 2005, Yirmiya & Shaked 2005]. In several studies rates of 10%-45% for social impairment, aloofness, shyness, and pragmatic language impairment were present in at least one parent of children with autism spectrum disorders [Freitag 2007]. Cederlund & Gillberg [2004] reported that 70% of probands with autism of unknown cause studied had a first- or second-degree relative with autistic symptoms, and that 15% had fathers with Asperger syndrome.

Sibs of a proband. The empiric aggregate risk to sibs of individuals with autism of unknown cause varies across studies but is generally considered to range from 5% to 10% for autism and 10% to 15% for milder symptoms, including language, social, and psychiatric disorders [Bolton et al 1994, Lauritsen et al 2005, Miles et al 2005, Landa 2008, Selkirk et al 2009]. For families with two or more affected children, the recurrence risk approaches 35% [Ritvo et al 1989]. No recurrence risk data are available for families who have one autistic child plus another child or relative with mild autistic symptoms. Therefore, the amount of weight to put on mild autistic symptoms in siblings, parents, and other relatives when estimating recurrence risk for families is unknown.

*

Essential autism
o

Male sibs (brothers) of a proband with essential autism have a 7% risk for autism and an additional 7% risk for milder autism spectrum symptoms [Miles et al 2005].
o

Female sibs (sisters) of a proband with essential autism have a 1% risk for autism. The risk for milder autism spectrum disorder is unknown [Miles et al 2005].
*

Complex autism. The recurrence risk to sibs of a proband with complex autism is 1% for autism and an additional 2% for milder autism spectrum symptoms [Miles et al 2005].

Offspring of a proband. No data are available.
Related Genetic Counseling Issues

DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals.

Prenatal Testing

Prenatal testing for families at risk of having a child with a chromosomal disorder, copy number variant, or a single gene disorder known to be associated with autism may be available for the specific etiology; search the GeneTests Laboratory Directory for information on the specific syndrome to asses the availability of prenatal testing.

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease in an individual diagnosed with autism, the following evaluations are recommended:

*

Medical evaluation. A complete medical evaluation and review of systems to assess growth and identify health problems that may interfere with optimum development and progress. In particular, note:
o

Sleep. Problems falling asleep, early or mid-night wakening and parasomnias are common. Routine sleep hygiene recommendations, search for underlying physical causes, and referral to sleep specialists are warranted. Medications should be delayed until underlying causes are ruled out.
o

Gastrointestinal symptoms including both constipation and diarrhea are common in autism and respond to medical management. Referral to a gastroenterologist is recommended if symptoms are chronic
o

Obesity. Children with autism are often treated with medications, such as atypical antipsychotics which can increase hunger and lead to weight gain. In addition, many children and teenagers are physically inactive, preferring the computer and video games.
o

Hearing evaluation
*

Neurologic dysfunction. Signs of seizures, focal neurologic deficits
*

Cognitive testing. A cognitive assessment by a psychologist experienced in autism evaluations; however, since IQ scores may change especially in young children it is recommended that children be retested every three years. This is usually done through the school system and those records should be obtained.
*

Language/communication assessment. Assess whether the child is functionally verbal or only has occasional words or echolalia. Language assessment by a trained speech/language pathologist with access to training modalities and communication modalities such as Picture Exchange Communication System (PECS). Even children who appear functionally verbal generally need ongoing speech and language therapy to gain skills in pragmatics and social and receptive communication.
*

Behavioral evaluation. Assess for repetitive behaviors, aggression, self-injurious behavior, mood, and anxiety.
*

Educational programs and interventions. Assess adequacy and progress.
*

Family functioning, including financial resources including qualifications for Medicaid, Social Security Supplemental Income, and other state specific programs

Treatment of Manifestations

Optimally, an autism intervention program uses an experienced team of medical, behavioral, and educational specialists. Working with an established team is easiest for families; however, many physicians and families have successfully assembled programs that work well.

Early diagnosis and early intensive behavioral therapy are essential to an optimal outcome. Two comprehensive treatment reviews are Educating Children with Autism [Lord et al 2001] and Management of Children with Autism Spectrum Disorders from the American Academy of Pediatrics [Myers & Johnson 2007] are both available online. Also recommended is an evaluation of single-subject studies to determine which educational practices meet the criteria of "evidence-based practice" [Odom et al 2003].
Behavioral and Educational Therapy

Many interventions have been developed for children and youth with autism in the past 20 years. The effects of some of these interventions have been documented in peer-reviewed published research. Other interventions, although marketed to parents, have not been rigorously tested. The goal for practitioners is to prescribe and (as applicable) use scientifically based practices (SBP) when developing and implementing intervention plans. SBPs are those practices that have a substantial research base and are shown to be effective with children and youth with autism. Simpson [2005] provided a synthesis of autism intervention research and assigned classifications based on the level of empirical support. The classification categories are: scientifically based practice, promising practice, limited supporting information for practice, and not recommended. The interventions discussed below are considered either scientifically based or promising practices. The majority of interventions with empirical evidence of efficacy use structured behavioral and educational approaches to teach children to comprehend and use language, attend to their environment, imitate others, interact socially, and play appropriately with toys. Features include a functional approach to behavior problems and predictable and routine classroom and home arrangements with planned transitions between activities and environments.
Universal Support and Skill-Based Strategies

Environmental supports are changes to the environment that help the individual with ASD be more successful without requiring acquisition of new skills. Often these supports serve as a foundation for other targeted practices. These supports typically provide physical organization of the work space, task organization, clarity of transitions, and visual schedules/strategies.

The Treatment and Education of Autistic and related Communication-handicapped Children (TEACCH) program is a scientific-based example of a practice designed to increase students’ independence through physical and task organization [Mesibov et al 2004]. The intervention is designed to create environments (most typically classrooms and related learning settings) that provide clear expectations, structure, and predictability and reduce tendencies by children with ASD to over or under attend to stimuli through the use of physical structures. Work stations are created with clear task areas, break areas, task cues to provide support for sequencing of steps, materials and behavior expectations. Many materials and areas are color coded and supplemented with visuals, either written or pictorial. Although TEACCH procedures are more frequently used in school settings, many practitioners incorporate many of these structures into community and home environments (e.g., a bin of preferred items with visual play cues for children to use while mom is cooking). Another frequently used strategy is Visual Schedules which are designed to increase predictability and reduce anxiety. Visual representations of tasks or events, including their sequence, duration, temporal distance, cause and effect without an overall reliance on auditory processing is beneficial to learners with ASD [Mesibov & Howley 2003]. Visual strategies alone do not teach communication and should not be confused with promising formalized scientific practice strategies that use visual representations. See Communication in this section.

Applied Behavior Analysis (ABA) is an intervention based on the use of behavioral principles to change, reduce, or increase behaviors. It incorporates high levels of environmental support and uses them to target skill acquisition across multiple developmental domains. ABA has no age boundaries and teaches target behaviors coupled with specific processes for responses to increase (reinforce) or decrease behaviors. ABA programs are based on repeated analyses of an individual’s strengths related to environmental functioning [Alberto & Troutman 2008]. This approach is referred to as “functional analysis” and is an integral component of the federal laws that regulate the delivery of special education services [ILIAD 2004 (Individuals with Disabilities Education Act)]. Functional analysis seeks to identify the purpose for which an individual exhibits a maladaptive or prosocial behavior. This allows development of individualized and effective environmental supports and skill-based techniques to teach new behaviors. ABA, including functional analysis, is the intervention technique with the strongest research base supporting its effectiveness in both ASD and non-ASD populations.

A number of evidence-based or emerging practices, including Discrete Trial Teaching and Pivotal Response Training, are based on ABA principles. One such technique that is expected to emerge shortly as scientific is Incidental Teaching, which uses basic concepts of ABA in the child’s natural environment [McGee et al 1999, Peterson et al 2005].

Incidental Teaching is implemented around the child’s interests and typical activities, using natural supports such as favorite toys, people or foods as the basis for teaching new skills. To date, research has demonstrated success across all levels of cognitive and ASD symptomology, and most age ranges, especially in early childhood [McGee et al 1999]. Skills acquired through Incidental Teaching are considered more generalizable to day-to-day functioning when compared to methods like discrete trial training, which breaks down single tasks and teaches them separately [Lord et al 2001, Simpson et al 2005].

Individuals with average intelligence or mild cognitive disability often benefit from a more cognitive processing strategy called Cognitive Behavior Intervention or cognitive learning strategies [Bauminger 2002, Solomon et al 2004, Webb et al 2004, Tse et al 2007].

Cognitive Behavior Intervention, categorized as a promising practice for ASD, teaches problem solving schemas using systematic application of environmental supports, rules, and principles which lead to self-management and behavior regulation. It is anticipated as more research is being published that this strategy will soon be moved to scientific-based status for ASD. This practice is particularly helpful for teaching self-management skills and regulating one’s own behavior.

Sensory Integration (SI) is the organization and processing of sensory information for specific functional use. Proponents of sensory integration therapy view the aberrant behavior of children with autism as an attempt to establish an internal state of equilibrium. Scientific theory exists for this view, but little empiric data support its use. It is currently rated a promising practice by Simpson and colleagues based on a growing body of literature [Simpson et al 2005]. Despite a lack of specific data to support its use for ASD, caregivers at home and in the classroom report decreased hyperactivity, inattention, and self-stimulation following SI therapy. Although the program should be designed by an occupational therapist with training in sensory integration, many of the techniques, including brushing, use of weighted vests, swinging, and jumping are easily adapted to home and school use.

Communication. Communication impairment is one of the defining features of autism. Effective interventions to teach language and communication are also based on the principals of applied behavior analysis (ABA) and vary by the extent to which teaching is integrated into the child’s normal activities. At one end of the spectrum is discrete trial training that consists of intensive, rigidly structured, adult-directed, one-on-one interventions [Prizant et al 2000]. Debate exists about the appropriateness and ultimate success of teaching language skills outside of the contexts in which language is used. However, programs that use a discrete-trial training format are supported by published data and are rated as evidence-based practice [Heflin & Alaimo 2007]. Principles of ABA are also used in interventions in which instruction is embedded in home and classroom routines and the interaction is either shared or child directed. For example, Pivotal Response Training, Incidental Teaching, and the Picture Exchange Communication System are three naturalistic interventions considered to be scientifically based or a promising practice [Simpson 2005]. These naturalistic interventions can be used at school or home, and although the primary focus is language, they also target cognitive, play, and social communication skills.

Pivotal Response Training (PRT) [Koegel & Kern Koegel 2006], a scientifically based practice [Simpson et al 2005], was developed to teach skills and behaviors that enhance the child’s ability to learn and maintain new behaviors by natural consequences in typical environments. The main components of PRT are child choice, interspersing new tasks with those already mastered, reinforcing child attempts, gaining the child’s attention before giving directions, and teaching with multiple cues. PRT uses the child’s preferred activities and interests to provide motivation for learning new skills. Incidental teaching [Kaiser 2000], a promising practice [Simpson et al 2005] uses naturally occurring routines and activities, including play activities to encourage child initiations, particularly child requests. Adults reinforce the child’s language by providing access to requested activities or objects, modeling appropriate language and building turn-taking routines. Because language is taught in context, incidental teaching can be used to teach a variety of skills.

Picture Exchange Communication System (PECS) [Bondy et al 2004], a promising practice [Simpson et al 2005], was developed as a functional communication system for nonverbal children. With PECS, children use visual representations to request a desired object or activity. Children can also be taught to use the visual representations to comment on objects or activities of interest. Children progress through six phases of training that include exchanging a picture for an object or activity, making choices among pictures, and answering questions. Although not the original intent, many young children who use PECS do learn to talk.

Social. Individuals with autism experience social deficits that inhibit their ability to make and sustain friendships and navigate complex social environments. In social situations, these individuals generally lack the skills to initiate social interactions and fail to respond appropriately, leading to common descriptors such as socially awkward, self-centered, or inflexible. They also do not pick up nonverbal social cues and social prompts, and tend to display socially unacceptable behavior [Myles & Simpson 2002]. Without effective and targeted intervention, these deficits significantly affect long-term prognosis [Howlin & Karpf 2004].

Young children and those more affected by autism symptoms may benefit from play-oriented strategies, which are particularly effective for developing skills in turn-taking, sharing functional communication, and waiting, all of which are essential for effective social interactions. Play-based interventions such as Integrated Play Group and Milieu Teaching target individualized goals for the child and are distinct from ‘play therapy,’ which is often not effective for children with ASD, due in part to the emphasis of symbolic representation [Wolfberg 1999]. Learning Experiences: An Alternative Program for Preschoolers and Parents (LEAP) is a scientifically based intervention specifically focusing on social skills that generally takes place in a preschool setting but can also be implemented at home [Strain & Hoyson 2000].

Similar to the cognitive behavior interventions is a group of promising practices categorized as Social Decision Making Strategies [Simpson et al 2005]. These practices provide steps for making decisions, generating alternatives and understanding appropriate solutions in social situations. Examples include: social autopsies (deconstructing social situations that went wrong), stop-plot-go-so (determining the ‘who,’ ‘what,’ ‘when,’ ‘where,’ and ‘then what’ of social situations) and ‘stop,’ ‘observe,’ ‘deliberate,’ and ‘act’ (SODA) [Myles & Simpson 2003].

Another promising practice is Social Stories™, which is used to increase appropriate behavior and decrease problem behavior by explaining social situations in ways that are understandable to the student [Gray 2000]. The assumption is that problem behavior is caused by lack of understanding of what is expected or what is going to happen next; social stories build understanding, allowing the student to behave appropriately. Social stories are inexpensive to create and take much less time and expertise to implement than other interventions. Social Stories have been used successfully with children with ASD across a continuum of ages and abilities. Some variations are called Power Cards and Comic Scripts [Gagnon 2001].

Medical management. Although most children with autism are healthy, evidence is mounting that medical disorders have a significant effect on behaviors, level of functioning, and response to educational therapies. Sensory issues including a blunted pain response, inability to tell others when they are uncomfortable, and poor tolerance of medical evaluations can lead to suboptimal medical care. Emerging areas of research include gastrointestinal, feeding, sleep, metabolic, and pain disorders [Linday et al 2001, Bradley et al 2004, Polimeni et al 2005]. Evidence of gastroesophogeal reflux causing insomnia and self-injurious behaviors are compelling and indicate that physicians need to have a high index of suspicion, especially with unexplained behavioral exacerbations, and to provide the same level of medical intervention as in typically developing children. The Autism Treatment Network (ATN) is a consortium of 15 autism treatment center which was formed with support of Autism Speaks and the National Institutes of Health to develop a medically based approach to the diagnosis and treatment of individuals with autism. Algorithms for the initial assessment of children with autism as well as guidelines for continued medical care and surveillance are being developed and can be accessed through the Web site as well as through planned publications.

The use of medications has increased as newer medications, especially the atypical antipsychotics, which affect both the serotonin and dopamine systems, and serotonin reuptake inhibitors (SRIs), which modulate the serotonin system, have been studied in children. In 1997, the National Institute of Mental Health formed the Research Units on Pediatric Psychopharmacology (RUPP) Autism Network to investigate the safety and efficacy of drugs for treating the behaviors associated with autism. Reports from that consortium have provided authoritative reviews of the pharmacotherapy of autism [McDougle et al 2005, Posey et al 2008]. The conclusions:

*

No medications are autism specific. Medications should be selected to ameliorate a specific symptom such as aggressive or self-injurious behavior, agitation, anxiety, poor sleep and repetitive or stereotypic behaviors that interfere with learning and social interactions [Bodfish 2004, McDougle et al 2005].
*

A medication that alleviates one maladaptive behavior, such as aggression or hyperactivity, may have no effect on core autistic symptoms.
*

Marked differences exist in the efficacy and side effects of drugs in adults vs children.
*

Individual antipsychotic medications within the same class may differ with respect to their potency and side effect profile.
*

Affected individuals may respond differently to the same medication. The response to a medication may reflect genetic differences between individuals, the waxing and waning of the behaviors over time, the progression of the disorder, and/or placebo effect.
*

Medication management should be integrated into a family centered, multi-modal behavioral and educational program.

In the treatment of the catatonia syndrome, antipsychotic medications are contraindicated and treatment with benzodiazepines and electroconvulsive therapy has been helpful [Wing & Shah 2000, Billstedt et al 2005, Kakooza-Mwesige et al 2008].

Alternative therapies. Complementary and alternative medical (CAM) treatments are commonly used for children with autism [Hanson et al 2007]. Levy & Hyman [2008] reviewed the evidence supporting the most frequently used treatments, including categories of mind-body medicine, energy medicine, and biologically based, manipulative, and body-based practices. Clinical providers need to understand the evidence for efficacy (or lack thereof) and potential side effects.

Though parents generally consider CAM practices a safe alternative to prescribed medications, most treatments have not been adequately studied and do not have evidence to support their use. Some CAM practices, such as administration of secretin, facilitated communication, auditory training, have evidence to reject their use. A few CAM practices such as administration of melatonin have emerging evidence to support their use.
Surveillance

Children with autism should be followed at least annually to monitor health, educational, language, and behavioral progress. An update of the areas covered in the initial evaluation is recommended. Identification of specific complications is a targeted goal of the Autism Treatment Network, which will be following a large cohort of individuals into adulthood. Since diagnostic and treatment recommendations will continue to be revised, periodic diagnostic reappraisals by a medical geneticist or an autism center are recommended. As more individuals receive etiologic diagnoses, diagnosis-specific surveillance profiles will be developed.

Complications for which evidence supports routine monitoring include:

*

Seizures
*

Sleep disturbances
*

Feeding problems
*

Gastrointestinal symptoms
*

Medication-specific side effects
*

Mood and psychiatric dysfunction
*

Medication-induced complications. All children on medications should be closely monitored for medication side effects.

Agents/Circumstances to Avoid

A hands-off or wait-and-see approach should be avoided because children with autism are unlikely to be able to navigate the social world and develop reciprocal communication skills without intensive therapeutic intervention.

Families need to be cautioned about unproven and potentially dangerous “alternative therapies” that may be promoted either by well-meaning but naïve people or by unscrupulous groups, individuals, or clinics.
Testing of Relatives at Risk

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.
Therapies Under Investigation

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions. Note: There may not be clinical trials for this disorder.
Other

Genetics clinics, staffed by genetics professionals, provide information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

See Consumer Resources for disease-specific and/or umbrella support organizations for this disorder. These organizations have been established for individuals and families to provide information, support, and contact with other affected individuals.

Resources

See Consumer Resources for disease-specific and/or umbrella support organizations for this disorder. These organizations have been established for individuals and families to provide information, support, and contact with other affected individuals. GeneTests provides information about selected organizations and resources for the benefit of the reader; GeneTests is not responsible for information provided by other organizations.—ED.

References

Medical Genetic Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page link.
Literature Cited

1. Alarcón M, Abrahams BS, Stone JL, Duvall JA, Perederiy JV, Bomar JM, Sebat J, Wigler M, Martin CL, Ledbetter DH, Nelson SF, Cantor RM, Geschwind DH. Linkage, association, and gene-expression analyses identify CNTNAP2 as an autism-susceptibility gene. Am J Hum Genet. 2008;82:150–9. [PubMed: 18179893]
2. Alberto PA, Troutman AC. Applied Behavior Analysis for Teachers. Upper Saddle River, NJ: Merril/Prentice Hall; 2008.
3. Alvarez Retuerto AI, Cantor RM, Gleeson JG, Ustaszewska A, Schackwitz WS, Pennacchio LA, Geschwind DH. Association of common variants in the Joubert syndrome gene (AHI1) with autism. Hum Mol Genet. 2008;17:3887–96. [PubMed: 18782849]
4. Aman MG, Singh NN, Stewart AW, Field CJ. The aberrant behavior checklist: a behavior rating scale for the assessment of treatment effects. Am J Ment Defic. 1985;89:485–91. [PubMed: 3993694]
5. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington DC: American Psychiatric Association; 2000.
6. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat Genet. 1999;23:185–8. [PubMed: 10508514]
7. Arking DE, Cutler DJ, Brune CW, Teslovich TM, West K, Ikeda M, Rea A, Guy M, Lin S, Cook EH, Chakravarti A. A common genetic variant in the neurexin superfamily member CNTNAP2 increases familial risk of autism. Am J Hum Genet. 2008;82:160–4. [PubMed: 18179894]
8. Arvidsson T, Danielsson B, Forsberg P, Gillberg C, Johansson M, Kjellgren G. Autism in 3-6-year-old children in a suburb of Goteborg, Sweden. Autism. 1997;1:163–73.
9. Asano E, Chugani DC, Muzik O, Behen M, Janisse J, Rothermel R, Mangner TJ, Chakraborty PK, Chugani HT. Autism in tuberous sclerosis complex is related to both cortical and subcortical dysfunction. Neurology. 2001;57:1269–77. [PubMed: 11591847]
10. Asperger H. Die Autistischen Psychopathen in Kindersalter. Cambridge, UK: Cambridge University Press; 1944.
11. Atladottir HO, Parner ET, Schendel D, Dalsgaard S, Thomsen PH, Thorsen P. Time trends in reported diagnoses of childhood neuropsychiatric disorders: a Danish cohort study. Arch Pediatr Adolesc Med. 2007;161:193–8. [PubMed: 17283306]
12. Attwood T. Asperger's Diagnostic Assessment. Video recording. 2004. Arlington, TX: Future Horizons.
13. Autism and Developmental Disabilities Monitoring Network; Autism and Developmental Disabilities Monitoring Network Surveillance Year 2006 Principal Investigators; Centers for Disease Control and Prevention (CDC). Prevalence of autism spectrum disorders - Autism and Developmental Disabilities Monitoring Network, United States, 2006. MMWR Surveill Summ. 2009;58:1–20.
14. Baieli S, Pavone L, Meli C, Fiumara A, Coleman M. Autism and phenylketonuria. J Autism Dev Disord. 2003;33:201–4. [PubMed: 12757360]
15. Baird G, Charman T, Baron-Cohen S, Cox A, Swettenham J, Wheelwright S, Drew A. A screening instrument for autism at 18 months of age: a 6-year follow-up study. J Am Acad Child Adolesc Psychiatry. 2000;39:694–702. [PubMed: 10846303]
16. Baird G, Simonoff E, Pickles A, Chandler S, Loucas T, Meldrum D, Charman T. Prevalence of disorders of the autism spectrum in a population cohort of children in South Thames: the Special Needs and Autism Project (SNAP). Lancet. 2006;368:210–5. [PubMed: 16844490]
17. Baker P, Piven J, Sato Y. Autism and tuberous sclerosis complex: prevalence and clinical features. J Autism Dev Disord. 1998;28:279–85. [PubMed: 9711484]
18. Bakkaloglu B, O'Roak BJ, Louvi A, Gupta AR, Abelson JF, Morgan TM, Chawarska K, Klin A, Ercan-Sencicek AG, Stillman AA, Tanriover G, Abrahams BS, Duvall JA, Robbins EM, Geschwind DH, Biederer T, Gunel M, Lifton RP, State MW. Molecular cytogenetic analysis and resequencing of contactin associated protein-like 2 in autism spectrum disorders. Am J Hum Genet. 2008;82:165–73. [PubMed: 18179895]
19. Bandim JM, Ventura LO, Miller MT, Almeida HC, Costa AE. Autism and Mobius sequence: an exploratory study of children in northeastern Brazil. Arq Neuropsiquiatr. 2003;61:181–5. [PubMed: 12806493]
20. Barrett CF, Tsien RW. The Timothy syndrome mutation differentially affects voltage- and calcium-dependent inactivation of CaV1.2 L-type calcium channels. Proc Natl Acad Sci USA. 2008;105:2157–62. [PubMed: 18250309]
21. Battaglia A, Carey JC. Etiologic yield of autistic spectrum disorders: a prospective study. Am J Med Genet C Semin Med Genet. 2006;142C:3–7. [PubMed: 16419094]
22. Bauminger N. The facilitation of social-emotional understanding and social interaction in high-functioning children with autism: intervention outcomes. J Autism Dev Disord. 2002;32:283–98. [PubMed: 12199133]
23. Ben Shachar S, Lanpher B, German JR, Qasaymeh M, Potocki L, Nagamani SC, Franco LM, Malphrus A, Bottenfield GW, Spence JE, Amato S, Rousseau JA, Moghaddam B, Skinner C, Skinner SA, Bernes S, Armstrong N, Shinawi M, Stankiewicz P, Patel A, Cheung SW, Lupski JR, Beaudet AL, Sahoo T. Microdeletion 15q13.3: a locus with incomplete penetrance for autism, mental retardation, and psychiatric disorders. J Med Genet. 2009;46:382–8. [PubMed: 19289393]
24. Benayed R, Gharani N, Rossman I, Mancuso V, Lazar G, Kamdar S, Bruse SE, Tischfield S, Smith BJ, Zimmerman RA, Dicicco-Bloom E, Brzustowicz LM, Millonig JH. Support for the homeobox transcription factor gene ENGRAILED 2 as an autism spectrum disorder susceptibility locus. Am J Hum Genet. 2005;77:851–68. [PubMed: 16252243]
25. Berg JS, Brunetti-Pierri N, Peters SU, Kang SH, Fong CT, Salamone J, Freedenberg D, Hannig VL, Prock LA, Miller DT, Raffalli P, Harris DJ, Erickson RP, Cunniff C, Clark GD, Blazo MA, Peiffer DA, Gunderson KL, Sahoo T, Patel A, Lupski JR, Beaudet AL, Cheung SW. Speech delay and autism spectrum behaviors are frequently associated with duplication of the 7q11.23 Williams-Beuren syndrome region. Genet Med. 2007;9:427–41. [PubMed: 17666889]
26. Billstedt E, Gillberg IC, Gillberg C. Autism after adolescence: population-based 13- to 22-year follow-up study of 120 individuals with autism diagnosed in childhood. J Autism Dev Disord. 2005;35:351–60. [PubMed: 16119476]
27. Bird A. The methyl-CpG-binding protein MeCP2 and neurological disease. Biochem Soc Trans. 2008;36:575–83. [PubMed: 18631120]
28. Boddaert N, Zilbovicius M, Philipe A, Robel L, Bourgeois M, Barthelemy C, Seidenwurm D, Meresse I, Laurier L, Desguerre I, Bahi-Buisson N, Brunelle F, Munnich A, Samson Y, Mouren MC, Chabane N. MRI findings in 77 children with non-syndromic autistic disorder. PLoS ONE. 2009;4:e4415. [PubMed: 19204795]
29. Bodfish JW. Treating the core features of autism: are we there yet? Ment Retard Dev Disabil Res Rev. 2004;10:318–26. [PubMed: 15666340]
30. Bolton P, Macdonald H, Pickles A, Rios P, Goode S, Crowson M, Bailey A, Rutter M. A case-control family history study of autism. J Child Psychol Psychiatry. 1994;35:877–900. [PubMed: 7962246]
31. Bolton PF. Neuroepileptic correlates of autistic symptomatology in tuberous sclerosis. Ment Retard Dev Disabil Res Rev. 2004;10:126–31. [PubMed: 15362169]
32. Bolton PF, Pickles A, Murphy M, Rutter M. Autism, affective and other psychiatric disorders: patterns of familial aggregation. Psychol Med. 1998;28:385–95. [PubMed: 9572095]
33. Bonati MT, Russo S, Finelli P, Valsecchi MR, Cogliati F, Cavalleri F, Roberts W, Elia M, Larizza L. Evaluation of autism traits in Angelman syndrome: a resource to unfold autism genes. Neurogenetics. 2007;8:169–78. [PubMed: 17415598]
34. Bondy A, Tincani M, Frost L. Multiply controlled verbal operants: an analysis and extension to the picture exchange communication system. Behavior Analyst. 2004;27:247–61.
35. Bonora E, Beyer KS, Lamb JA, Parr JR, Klauck SM, Benner A, Paolucci M, Abbott A, Ragoussis I, Poustka A, Bailey AJ, Monaco AP. Analysis of reelin as a candidate gene for autism. Mol Psychiatry. 2003;8:885–92. [PubMed: 14515139]
36. Bonora E, Lamb JA, Barnby G, Sykes N, Moberly T, Beyer KS, Klauck SM, Poustka F, Bacchelli E, Blasi F, Maestrini E, Battaglia A, Haracopos D, Pedersen L, Isager T, Eriksen G, Viskum B, Sorensen EU, Brondum-Nielsen K, Cotterill R, Engeland H, Jonge M, Kemner C, Steggehuis K, Scherpenisse M, Rutter M, Bolton PF, Parr JR, Poustka A, Bailey AJ, Monaco AP. Mutation screening and association analysis of six candidate genes for autism on chromosome 7q. Eur J Hum Genet. 2005;13:198–207. [PubMed: 15523497]
37. Borgatti R, Piccinelli P, Passoni D, Raggi E, Ferrarese C. Pervasive developmental disorders and GABAergic system in patients with inverted duplicated chromosome 15. J Child Neurol. 2001;16:911–4. [PubMed: 11785506]
38. Bradley EA, Summers JA, Wood HL, Bryson SE. Comparing rates of psychiatric and behavior disorders in adolescents and young adults with severe mental retardation with and without autism. J Autism Dev Disord. 2004;34:151–61. [PubMed: 15162934]
39. Briegel W, Schimek M, Kamp-Becker I, Hofmann C, Schwab KO. Autism spectrum disorders in children and adolescents with Moebius sequence. Eur Child Adolesc Psychiatry. 2009;18:515–9. [PubMed: 19255803]
40. Bromley RL, Mawer G, Clayton-Smith J, Baker GA. Autism spectrum disorders following in utero exposure to antiepileptic drugs. Neurology. 2008;71:1923–4. [PubMed: 19047565]
41. Butler MG, Dasouki MJ, Zhou XP, Talebizadeh Z, Brown M, Takahashi TN, Miles JH, Wang CH, Stratton R, Pilarski R, Eng C. Subset of individuals with autism spectrum disorders and extreme macrocephaly associated with germline PTEN tumour suppressor gene mutations. J Med Genet. 2005;42:318–21. [PubMed: 15805158]
42. Buxbaum JD, Cai G, Chaste P, Nygren G, Goldsmith J, Reichert J, Anckarsater H, Rastam M, Smith CJ, Silverman JM, Hollander E, Leboyer M, Gillberg C, Verloes A, Betancur C. Mutation screening of the PTEN gene in patients with autism spectrum disorders and macrocephaly. Am J Med Genet B Neuropsychiatr Genet. 2007a;144B:484–91. [PubMed: 17427195]
43. Buxbaum JD, Cai G, Nygren G, Chaste P, Delorme R, Goldsmith J, Rastam M, Silverman JM, Hollander E, Gillberg C, Leboyer M, Betancur C. Mutation analysis of the NSD1 gene in patients with autism spectrum disorders and macrocephaly. BMC Med Genet. 2007b;8:68. [PubMed: 18001468]
44. Buxbaum JD, Silverman JM, Smith CJ, Greenberg DA, Kilifarski M, Reichert J, Cook EH, Fang Y, Song CY, Vitale R. Association between a GABRB3 polymorphism and autism. Mol Psychiatry. 2002;7:311–6. [PubMed: 11920158]
45. Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R, Bravaccio C, Trillo S, Elia M, Schneider C, Melmed R, Sacco R, Persico AM, Levitt P. A genetic variant that disrupts MET transcription is associated with autism. Proc Natl Acad Sci U S A. 2006;103:16834–9. [PubMed: 17053076]
46. Cavagnaro AT. Autistic Spectrum Disorders: Changes in the California Caseload: An Update: 1987-2007. Sacramento, CA: California Department of Developmental Services. California Health and Human Services Agency. Available at www.dds.ca.gov (pdf). 2007. Accessed 4-10-10.
47. Cederlund M, Gillberg C. One hundred males with Asperger syndrome: a clinical study of background and associated factors. Dev Med Child Neurol. 2004;46:652–60. [PubMed: 15473168]
48. Chakrabarti S, Fombonne E. Pervasive developmental disorders in preschool children. JAMA. 2001;285:3093–9. [PubMed: 11427137]
49. Chakrabarti S, Fombonne E. Pervasive developmental disorders in preschool children: confirmation of high prevalence. Am J Psychiatry. 2005;162:1133–41. [PubMed: 15930062]
50. Chaste P, Nygren G, Anckarsater H, Rastam M, Coleman M, Leboyer M, Gillberg C, Betancur C. Mutation screening of the ARX gene in patients with autism. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:228–30. [PubMed: 17044103]
51. Chawarska K, Bearss K. Assessment of cognitive and adaptive skills. In: Chawarska K, Klin A, Volkmar FR, eds. Autism Spectrum Disorders in Infants and Toddlers: Diagnosis, Assessment, and Treatment. New York / London: The Guildford Press; 2008.
52. Chih B, Afridi SK, Clark L, Scheiffele P. Disorder-associated mutations lead to functional inactivation of neuroligins. Hum Mol Genet. 2004;13:1471–7. [PubMed: 15150161]
53. Cho IH, Yoo HJ, Park M, Lee YS, Kim SA. Family-based association study of 5-HTTLPR and the 5-HT2A receptor gene polymorphisms with autism spectrum disorder in Korean trios. Brain Res. 2007;1139:34–41. [PubMed: 17280648]
54. Christian SL, Brune CW, Sudi J, Kumar RA, Liu S, Karamohamed S, Badner JA, Matsui S, Conroy J, McQuaid D, Gergel J, Hatchwell E, Gilliam TC, Gershon ES, Nowak NJ, Dobyns WB, Cook EH. Novel submicroscopic chromosomal abnormalities detected in autism spectrum disorder. Biol Psychiatry. 2008;63:1111–7. [PubMed: 18374305]
55. Chubykin AA, Liu X, Comoletti D, Tsigelny I, Taylor P, Sudhof TC. Dissection of synapse induction by neuroligins: effect of a neuroligin mutation associated with autism. J Biol Chem. 2005;280:22365–74. [PubMed: 15797875]
56. Coleman M. Advances in autism research. Dev Med Child Neurol. 2005;47:148. [PubMed: 15739717]
57. Collins AL, Ma D, Whitehead PL, Martin ER, Wright HH, Abramson RK, Hussman JP, Haines JL, Cuccaro ML, Gilbert JR, Pericak-Vance MA. Investigation of autism and GABA receptor subunit genes in multiple ethnic groups. Neurogenetics. 2006;7:167–74. [PubMed: 16770606]
58. Conciatori M, Stodgell CJ, Hyman SL, O'Bara M, Militerni R, Bravaccio C, Trillo S, Montecchi F, Schneider C, Melmed R, Elia M, Crawford L, Spence SJ, Muscarella L, Guarnieri V, D'Agruma L, Quattrone A, Zelante L, Rabinowitz D, Pascucci T, Puglisi-Allegra S, Reichelt KL, Rodier PM, Persico AM. Association between the HOXA1 A218G polymorphism and increased head circumference in patients with autism. Biol Psychiatry. 2004;55:413–9. [PubMed: 14960295]
59. Connors SL, Crowell DE, Eberhart CG, Copeland J, Newschaffer CJ, Spence SJ, Zimmerman AW. beta2-adrenergic receptor activation and genetic polymorphisms in autism: data from dizygotic twins. J Child Neurol. 2005;20:876–84. [PubMed: 16417856]
60. Constantino JN, Abbacchi AM, Lavesser PD, Reed H, Givens L, Chiang L, Gray T, Gross M, Zhang Y, Todd RD. Developmental course of autistic social impairment in males. Dev Psychopathol. 2009;21:127–38. [PubMed: 19144226]
61. Cook EHJ, Courchesne RY, Cox NJ, Lord C, Gonen D, Guter SJ, Lincoln A, Nix K, Haas R, Leventhal BL, Courchesne E. Linkage-disequilibrium mapping of autistic disorder, with 15q11-13 markers. Am J Hum Genet. 1998;62:1077–83. [PubMed: 9545402]
62. Cornish K, Kogan C, Turk J, Manly T, James N, Mills A, Dalton A. The emerging fragile X premutation phenotype: evidence from the domain of social cognition. Brain Cogn. 2005;57:53–60. [PubMed: 15629215]
63. Correia C, Coutinho AM, Diogo L, Grazina M, Marques C, Miguel T, Ataide A, Almeida J, Borges L, Oliveira C, Oliveira G, Vicente AM. Brief report: High frequency of biochemical markers for mitochondrial dysfunction in autism: no association with the mitochondrial aspartate/glutamate carrier SLC25A12 gene. J Autism Dev Disord. 2006;36:1137–40. [PubMed: 17151801]
64. Cortesi M, Alfei E, Barale F, Fusar-Poli P. Linking autism, regression and Landau-Kleffner syndrome: integrative role of nerve growth factor. Med Hypotheses. 2007;68:1178–9. [PubMed: 17157996]
65. Coutinho AM, Oliveira G, Morgadinho T, Fesel C, Macedo TR, Bento C, Marques C, Ataide A, Miguel T, Borges L, Vicente AM. Variants of the serotonin transporter gene (SLC6A4) significantly contribute to hyperserotonemia in autism. Mol Psychiatry. 2004;9:264–71. [PubMed: 15094787]
66. de Vries PJ, Hunt A, Bolton PF. The psychopathologies of children and adolescents with tuberous sclerosis complex (TSC): a postal survey of UK families. Eur Child Adolesc Psychiatry. 2007;16:16–24. [PubMed: 17268883]
67. Delatycki MB, Danks A, Churchyard A, Zhou XP, Eng C. De novo germline PTEN mutation in a man with Lhermitte-Duclos disease which arose on the paternal chromosome and was transmitted to his child with polydactyly and Wormian bones. J Med Genet. 2003;40:e92. [PubMed: 12920084]
68. DeStefano F, Thompson WW. MMR vaccine and autism: an update of the scientific evidence. Expert Rev Vaccines. 2004;3:19–22. [PubMed: 14761240]
69. Devlin B, Bennett P, Dawson G, Figlewicz DA, Grigorenko EL, McMahon W, Minshew N, Pauls D, Smith M, Spence MA, Rodier PM, Stodgell C, Schellenberg GD. CPEA Genetics Network; Alleles of a reelin CGG repeat do not convey liability to autism in a sample from the CPEA network. Am J Med Genet B Neuropsychiatr Genet. 2004;126B:46–50. [PubMed: 15048647]
70. Devlin B, Cook EH, Coon H, Dawson G, Grigorenko EL, McMahon W, Minshew N, Pauls D, Smith M, Spence MA, Rodier PM, Stodgell C, Schellenberg GD. Autism and the serotonin transporter: the long and short of it. Mol Psychiatry. 2005;10:1110–6. [PubMed: 16103890]
71. Durand CM, Betancur C, Boeckers TM, Bockmann J, Chaste P, Fauchereau F, Nygren G, Rastam M, Gillberg IC, Anckarsater H, Sponheim E, Goubran-Botros H, Delorme R, Chabane N, Mouren-Simeoni MC, de Mas P, Bieth E, Roge B, Heron D, Burglen L, Gillberg C, Leboyer M, Bourgeron T. Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders. Nat Genet. 2007;39:25–7. [PubMed: 17173049]
72. Dykens EM, Sutcliffe JS, Levitt P. Autism and 15q11-q13 disorders: behavioral, genetic, and pathophysiological issues. Ment Retard Dev Disabil Res Rev. 2004;10:284–91. [PubMed: 15666333]
73. Ehlers S, Gillberg C, Wing L. A screening questionnaire for Asperger syndrome and other high-functioning autism spectrum disorders in school age children. J Autism Dev Disord. 1999;29:129–41. [PubMed: 10382133]
74. Farley MA, McMahon WM, Fombonne E, Jenson WR, Miller J, Gardner M, Block H, Pingree CB, Ritvo ER, Ritvo RA, Coon H. Twenty-year outcome for individuals with autism and average or near-average cognitive abilities. Autism Res. 2009:109–18. [PubMed: 19455645]
75. Farzin F, Perry H, Hessl D, Loesch D, Cohen J, Bacalman S, Gane L, Tassone F, Hagerman P, Hagerman R. Autism spectrum disorders and attention-deficit/hyperactivity disorder in boys with the fragile X premutation. J Dev Behav Pediatr. 2006;27:S137–44. [PubMed: 16685180]
76. Feng J, Schroer R, Yan J, Song W, Yang C, Bockholt A, Cook EH, Skinner C, Schwartz CE, Sommer SS. High frequency of neurexin 1beta signal peptide structural variants in patients with autism. Neurosci Lett. 2006;409:10–3. [PubMed: 17034946]
77. Fernandez T, Morgan T, Davis N, Klin A, Morris A, Farhi A, Lifton RP, State MW. Disruption of contactin 4 (CNTN4) results in developmental delay and other features of 3p deletion syndrome. Am J Hum Genet. 2004;74:1286–93. [PubMed: 15106122]
78. Fernandez TV, Garcia-Gonzalez IJ, Mason CE, Hernandez-Zaragoza G, Ledezma-Rodriguez VC, Anguiano-Alvarez VM. Molecular characterization of a patient with 3p deletion syndrome and a review of the literature. Am J Med Genet A. 2008;146A:2746–52. [PubMed: 18837054]
79. Filipek PA, Accardo PJ, Ashwal S, Baranek GT, Cook EH, Dawson G, Gordon B, Gravel JS, Johnson CP, Kallen RJ, Levy SE, Minshew NJ, Ozonoff S, Prizant BM, Rapin I, Rogers SJ, Stone WL, Teplin SW, Tuchman RF, Volkmar FR. Practice parameter: screening and diagnosis of autism: report of the Quality Standards Subcommittee of the American Academy of Neurology and the Child Neurology Society. Neurology. 2000;55:468–79. [PubMed: 10953176]
80. Filipek PA, Accardo PJ, Baranek GT, Cook EH, Dawson G, Gordon B, Gravel JS, Johnson CP, Kallen RJ, Levy SE, Minshew NJ, Ozonoff S, Prizant BM, Rapin I, Rogers SJ, Stone WL, Teplin S, Tuchman RF, Volkmar FR. The screening and diagnosis of autistic spectrum disorders. J Autism Dev Disord. 1999;29:439–84. [PubMed: 10638459]
81. Fombonne E. Is there an epidemic of autism? Pediatrics. 2001;107:411–2. [PubMed: 11158478]
82. Fombonne E, Bolton P, Prior J, Jordan H, Rutter M. A family study of autism: cognitive patterns and levels in parents and siblings. J Child Psychol Psychiatry. 1997a;38:667–83. [PubMed: 9315977]
83. Fombonne E, Du M, Cans C, Grandjean H. Autism and associated medical disorders in a French epidemiological survey. J Am Acad Child Adolesc Psychiatry. 1997b;36:1561–9. [PubMed: 9394941]
84. Fombonne E, Roge B, Claverie J, Courty S, Fremolle J. Microcephaly and macrocephaly in autism. J Autism Dev Disord. 1999;29:113–9. [PubMed: 10382131]
85. Fombonne E, Zakarian R, Bennett A, Meng L, McLean-Heywood D. Pervasive developmental disorders in Montreal, Quebec, Canada: prevalence and links with immunizations. Pediatrics. 2006;118:e139–e150. [PubMed: 16818529]
86. Fombonne E. Epidemiological studies of pervasive developmental disorders. In: Volkmar FR, Paul R, Klin A, Cohen DJ, eds. Handbook of Autism and Pervasive Developmental Disorders. Vol 1: Diagnosis, Developmental, Neurobiology and Behavior, 3rd ed. Hoboken, NJ: Wiley; 2005.
87. Freitag CM. The genetics of autistic disorders and its clinical relevance: a review of the literature. Mol Psychiatry. 2007;12:2–22. [PubMed: 17033636]
88. Frith U. 'Autistic psychopathy' in childhood. In Frith U, ed. Autism and Asperger Syndrome. Cambridge, UK: Cambridge University Press; 1991:37–92.
89. Gagnon E. Power Cards: Using Special Interests To Motivate Children and Youth with Asperger Syndrome and Autism. 2001.
90. Gallagher L, Hawi Z, Kearney G, Fitzgerald M, Gill M. No association between allelic variants of HOXA1/HOXB1 and autism. Am J Med Genet B Neuropsychiatr Genet. 2004;124B:64–7. [PubMed: 14681917]
91. Garber K. Neuroscience. Autism's cause may reside in abnormalities at the synapse. Science. 2007;317:190–1. [PubMed: 17626859]
92. Garnett MS, Atwood A. The Australian scale for Asperger's Syndrome. Asperger's Syndrome: A Guide for Parents and Professionals. London: Jessica Kingsley Publishers; 1997.
93. Gauthier J, Bonnel A, St-Onge J, Karemera L, Laurent S, Mottron L, Fombonne E, Joober R, Rouleau GA. NLGN3/NLGN4 gene mutations are not responsible for autism in the Quebec population. Am J Med Genet B Neuropsychiatr Genet. 2005;132B:74–5. [PubMed: 15389766]
94. Gauthier J, Joober R, Mottron L, Laurent S, Fuchs M, De Kimpe V, Rouleau GA. Mutation screening of FOXP2 in individuals diagnosed with autistic disorder. Am J Med Genet A. 2003;118A:172–5. [PubMed: 12655497]
95. Gernsbacher M, Dawson M, Goldsmith H. Three reasons not to believe in an autism epidemic. Curr Dir Psych Sci. 2005;14:55–8.
96. Geschwind DH, Sowinski J, Lord C, Iversen P, Shestack J, Jones P, Ducat L, Spence SJ. The autism genetic resource exchange: a resource for the study of autism and related neuropsychiatric conditions. Am J Hum Genet. 2001;69:463–6. [PubMed: 11452364]
97. Gharani N, Benayed R, Mancuso V, Brzustowicz LM, Millonig JH. Association of the homeobox transcription factor, ENGRAILED 2, 3, with autism spectrum disorder. Mol Psychiatry. 2004;9:474–84. [PubMed: 15024396]
98. Gillberg C. Chromosomal disorders and autism. J Autism Dev Disord. 1998;28:415–25. [PubMed: 9813777]
99. Gillberg C, Gillberg C, Rastam M, Wentz E. The Asperger syndrome (and high-functioning autism) Diagnostic Interview (ASDI): a preliminary study of a new structured clinical interview. Autism. 2001;5:57–66. [PubMed: 11708390]
100. Gilliam JE. Gilliam Autism Rating Scale (GARS). Austin, Texas: PRO-ED, Inc; 1995.
101. Glessner JT, Wang K, Cai G, Korvatska O, Kim CE, Wood S, Zhang H, Estes A, Brune CW, Bradfield JP, Imielinski M, Frackelton EC, Reichert J, Crawford EL, Munson J, Sleiman PM, Chiavacci R, Annaiah K, Thomas K, Hou C, Glaberson W, Flory J, Otieno F, Garris M, Soorya L, Klei L, Piven J, Meyer KJ, Anagnostou E, Sakurai T, Game RM, Rudd DS, Zurawiecki D, McDougle CJ, Davis LK, Miller J, Posey DJ, Michaels S, Kolevzon A, Silverman JM, Bernier R, Levy SE, Schultz RT, Dawson G, Owley T, McMahon WM, Wassink TH, Sweeney JA, Nurnberger JI, Coon H, Sutcliffe JS, Minshew NJ, Grant SF, Bucan M, Cook EH, Buxbaum JD, Devlin B, Schellenberg GD, Hakonarson H. Autism genome-wide copy number variation reveals ubiquitin and neuronal genes. Nature. 2009;459:569–73. [PubMed: 19404257]
102. Gong X, Jia M, Ruan Y, Shuang M, Liu J, Wu S, Guo Y, Yang J, Ling Y, Yang X, Zhang D. Association between the FOXP2 gene and autistic disorder in Chinese population. Am J Med Genet B Neuropsychiatr Genet. 2004;127B:113–6. [PubMed: 15108192]
103. Goodlin-Jones BL, Tassone F, Gane LW, Hagerman RJ. Autistic spectrum disorder and the fragile X premutation. J Dev Behav Pediatr. 2004;25:392–8. [PubMed: 15613987]
104. Gottesman II, Gould TD. The endophenotype concept in psychiatry: etymology and strategic intentions. Am J Psychiatry. 2003;160:636–45. [PubMed: 12668349]
105. Gray CA.The New Social Story Book. Arlington, TX: Future Horizons; 2000.
106. Grice DE, Buxbaum JD. The genetics of autism spectrum disorders. Neuromolecular Med. 2006;8:451–60. [PubMed: 17028369]
107. Gupta AR, State MW. Recent advances in the genetics of autism. Biol Psychiatry. 2007;61:429–37. [PubMed: 16996486]
108. Hagerman RJ, Rivera SM, Hagerman PJ. The Fragile X Family of Disorders: A Model for Autism and Targeted Treatments. Curr Pediatr Rev. 2008;4:40–52.
109. Handa V, Goldwater D, Stiles D, Cam M, Poy G, Kumari D, Usdin K. Long CGG-repeat tracts are toxic to human cells: implications for carriers of Fragile X premutation alleles. FEBS Lett. 2005;579:2702–8. [PubMed: 15862312]
110. Hanson E, Kalish LA, Bunce E, Curtis C, McDaniel S, Ware J, Petry J. Use of complementary and alternative medicine among children diagnosed with autism spectrum disorder. J Autism Dev Disord. 2007;37:628–36. [PubMed: 16977497]
111. Hara H. Autism and epilepsy: a retrospective follow-up study. Brain Dev. 2007;29:486–90. [PubMed: 17321709]
112. Harris SW, Hessl D, Goodlin-Jones B, Ferranti J, Bacalman S, Barbato I, Tassone F, Hagerman PJ, Herman H, Hagerman RJ. Autism profiles of males with fragile X syndrome. Am J Ment Retard. 2008;113:427–38. [PubMed: 19127654]
113. Heflin LJ, Alaimo DF. Students with Autism Spectrum Disorders: Effective Instructional Practices. Upper Saddle River, NJ: Pearson; 2007.
114. Hertz-Picciotto I, Delwiche L. The rise in autism and the role of age at diagnosis. Epidemiology. 2009;20:84–90. [PubMed: 19234401]
115. Hogart A, Wu D, LaSalle JM, Schanen NC. The comorbidity of autism with the genomic disorders of chromosome 15q11.2-q13. Neurobiol Dis. 2010;38(2):181–91. [PubMed: 18840528]
116. Hope CI, Sharp DM, Hemara-Wahanui A, Sissingh JI, Lundon P, Mitchell EA, Maw MA, Clover GM. Clinical manifestations of a unique X-linked retinal disorder in a large New Zealand family with a novel mutation in CACNA1F, the gene responsible for CSNB2. Clin Experiment Ophthalmol. 2005;33:129–36. [PubMed: 15807819]
117. Howlin P, Goode S, Hutton J, Rutter M. Adult outcome for children with autism. J Child Psychol Psychiatry. 2004;45:212–29. [PubMed: 14982237]
118. Howlin P, Karpf J. Using the social communication questionnaire to identify 'autistic spectrum' disorders associated with other genetic conditions: findings from a study of individuals with Cohen syndrome. Autism. 2004;8:175–82. [PubMed: 15165433]
119. Howlin P, Karpf J, Turk J. Behavioural characteristics and autistic features in individuals with Cohen Syndrome. Eur Child Adolesc Psychiatry. 2005;14:57–64. [PubMed: 15793684]
120. Howlin P, Mawhood L, Rutter M. Autism and developmental receptive language disorder--a follow-up comparison in early adult life. II: Social, behavioural, and psychiatric outcomes. J Child Psychol Psychiatry. 2000;41:561–78. [PubMed: 10946749]
121. Hutcheson HB, Olson LM, Bradford Y, Folstein SE, Santangelo SL, Sutcliffe JS, Haines JL. Examination of NRCAM, LRRN3, KIAA0716, and LAMB1 as autism candidate genes. BMC Med Genet. 2004;5:12. [PubMed: 15128462]
122. ILIAD. Individuals with Disabilities Education Improvement Act. Available at www.copyright.gov (pdf). 2004. Accessed 4-10-10.
123. Ingram JL, Stodgell CJ, Hyman SL, Figlewicz DA, Weitkamp LR, Rodier PM. Discovery of allelic variants of HOXA1 and HOXB1: genetic susceptibility to autism spectrum disorders. Teratology. 2000;62:393–405. [PubMed: 11091361]
124. Institute of Medicine. Immunization Safety Review: Vaccines and Autism. Washington, DC: The National Academies Press. Available at books.nap.edu. 2004. Accessed 4-10-10.
125. Jacob S, Brune CW, Carter CS, Leventhal BL, Lord C, Cook EH. Association of the oxytocin receptor gene (OXTR) in Caucasian children and adolescents with autism. Neurosci Lett. 2007;417:6–9. [PubMed: 17383819]
126. Jacquemont ML, Sanlaville D, Redon R, Raoul O, Cormier-Daire V, Lyonnet S, Amiel J, Le Merrer M, Heron D, de Blois MC, Prieur M, Vekemans M, Carter NP, Munnich A, Colleaux L, Philippe A. Array-based comparative genomic hybridisation identifies high frequency of cryptic chromosomal rearrangements in patients with syndromic autism spectrum disorders. J Med Genet. 2006;43:843–9. [PubMed: 16840569]
127. Jamain S, Quach H, Betancur C, Rastam M, Colineaux C, Gillberg IC, Soderstrom H, Giros B, Leboyer M, Gillberg C, Bourgeron T. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat Genet. 2003;34:27–9. [PubMed: 12669065]
128. Jansen VA, Stollenwerk N, Jensen HJ, Ramsay ME, Edmunds WJ, Rhodes CJ. Measles outbreaks in a population with declining vaccine uptake. Science. 2003;301:804. [PubMed: 12907792]
129. Jeste SS, Sahin M, Bolton P, Ploubidis GB, Humphrey A. Characterization of autism in young children with tuberous sclerosis complex. J Child Neurol. 2008;23:520–5. [PubMed: 18160549]
130. Jha P, Sheth D, Ghaziuddin M. Autism spectrum disorder and Klinefelter syndrome. Eur Child Adolesc Psychiatry. 2007;16:305–8. [PubMed: 17401614]
131. Jiang YH, Sahoo T, Michaelis RC, Bercovich D, Bressler J, Kashork CD, Liu Q, Shaffer LG, Schroer RJ, Stockton DW, Spielman RS, Stevenson RE, Beaudet AL. A mixed epigenetic/genetic model for oligogenic inheritance of autism with a limited role for UBE3A. Am J Med Genet A. 2004;131:1–10. [PubMed: 15389703]
132. Johansson M, Wentz E, Fernell E, Stromland K, Miller MT, Gillberg C. Autistic spectrum disorders in Mobius sequence: a comprehensive study of 25 individuals. Dev Med Child Neurol. 2001;43:338–45. [PubMed: 11368487]
133. Johnson CP, Myers SM. Identification and evaluation of children with autism spectrum disorders. Pediatrics. 2007;120:1183–215. [PubMed: 17967920]
134. Kaiser A. Teaching functional communication skills. In: Snell ME, Brown F, eds. Instruction for Students with Severe Disabilities. 5th ed. Columbus, OH: Merrill; 2000:453-91.
135. Kakooza-Mwesige A, Wachtel LE, Dhossche DM. Catatonia in autism: implications across the life span. Eur Child Adolesc Psychiatry. 2008;17:327–35. [PubMed: 18427869]
136. Kent L, Evans J, Paul M, Sharp M. Comorbidity of autistic spectrum disorders in children with Down syndrome. Dev Med Child Neurol. 1999;41:153–8. [PubMed: 10210247]
137. Kim HG, Kishikawa S, Higgins AW, Seong IS, Donovan DJ, Shen Y, Lally E, Weiss LA, Najm J, Kutsche K, Descartes M, Holt L, Braddock S, Troxell R, Kaplan L, Volkmar F, Klin A, Tsatsanis K, Harris DJ, Noens I, Pauls DL, Daly MJ, MacDonald ME, Morton CC, Quade BJ, Gusella JF. Disruption of neurexin 1 associated with autism spectrum disorder. Am J Hum Genet. 2008;82:199–207. [PubMed: 18179900]
138. Kim HL, Donnelly JH, Tournay AE, Book TM, Filipek P. Absence of seizures despite high prevalence of epileptiform EEG abnormalities in children with autism monitored in a tertiary care center. Epilepsia. 2006;47:394–8. [PubMed: 16499766]
139. Kleinman JM, Robins DL, Ventola PE, Pandey J, Boorstein HC, Esser EL, Wilson LB, Rosenthal MA, Sutera S, Verbalis AD, Barton M, Hodgson S, Green J, Dumont-Mathieu T, Volkmar F, Chawarska K, Klin A, Fein D. The modified checklist for autism in toddlers: a follow-up study investigating the early detection of autism spectrum disorders. J Autism Dev Disord. 2008;38:827–39. [PubMed: 17882539]
140. Klein-Tasman BP, Mervis CB, Lord C, Phillips KD. Socio-communicative deficits in young children with Williams syndrome: performance on the Autism Diagnostic Observation Schedule. Child Neuropsychol. 2007;13:444–67. [PubMed: 17805996]
141. Klin A, Pauls D, Schultz R, Volkmar F. Three diagnostic approaches to Asperger syndrome: implications for research. J Autism Dev Disord. 2005a;35:221–34. [PubMed: 15909408]
142. Klin A, Saulnier CA, Tsatsanis K, Volkmar FR.Clinical evaluation in autism spectrum disorders: Psychological assessment within a transdisciplinary framework. In: Volkmar FR, Paul R, Klin A, Cohen DJ, eds. Handbook of Autism and Pervasive Developmental Disorders. Vol 2: Assessment, Interventions, and Policy. 3rd ed. Hoboken, NJ: Wiley; 2005b.
143. Knoester M, Helmerhorst FM, van der Westerlaken LA, Walther FJ, Veen S. Matched follow-up study of 5 8-year-old ICSI singletons: child behaviour, parenting stress and child (health-related) quality of life. Hum Reprod. 2007;22:3098–107. [PubMed: 17905745]
144. Koegel RL, Kern Koegel L. Pivotal Response Treatments for Autism: Communication, Social, and Academic Development. Baltimore, MD: Brookes Publishing Company; 2006.
145. Kogan MD, Blumberg SJ, Schieve LA, Boyle CA, Perrin JM, Ghandour RM, Singh GK, Strickland BB, Trevathan E, van Dyck PC. Prevalence of parent-reported diagnosis of autism spectrum disorder among children in the US, 2007. Pediatrics. 2009;124:1395–403. [PubMed: 19805460]
146. Krebs MO, Betancur C, Leroy S, Bourdel MC, Gillberg C, Leboyer M. Absence of association between a polymorphic GGC repeat in the 5' untranslated region of the reelin gene and autism. Mol Psychiatry. 2002;7:801–4. [PubMed: 12192627]
147. Kumar RA, Karamohamed S, Sudi J, Conrad DF, Brune C, Badner JA, Gilliam TC, Nowak NJ, Cook EH, Dobyns WB, Christian SL. Recurrent 16p11.2 microdeletions in autism. Hum Mol Genet. 2008;17:628–38. [PubMed: 18156158]
148. Lainhart JE, Bigler ED, Bocian M, Coon H, Dinh E, Dawson G, Deutsch CK, Dunn M, Estes A, Tager-Flusberg H, Folstein S, Hepburn S, Hyman S, McMahon W, Minshew N, Munson J, Osann K, Ozonoff S, Rodier P, Rogers S, Sigman M, Spence MA, Stodgell CJ, Volkmar F. Head circumference and height in autism: a study by the Collaborative Program of Excellence in Autism. Am J Med Genet A. 2006;140:2257–74. [PubMed: 17022081]
149. Landa RJ. Diagnosis of autism spectrum disorders in the first 3 years of life. Nat Clin Pract Neurol. 2008;4:138–47. [PubMed: 18253102]
150. Laumonnier F, Bonnet-Brilhault F, Gomot M, Blanc R, David A, Moizard MP, Raynaud M, Ronce N, Lemonnier E, Calvas P, Laudier B, Chelly J, Fryns JP, Ropers HH, Hamel BC, Andres C, Barthelemy C, Moraine C, Briault S. X-linked mental retardation and autism are associated with a mutation in the NLGN4 gene, a member of the neuroligin family. Am J Hum Genet. 2004;74:552–7. [PubMed: 14963808]
151. Lauritsen MB, Pedersen CB, Mortensen PB. Effects of familial risk factors and place of birth on the risk of autism: a nationwide register-based study. J Child Psychol Psychiatry. 2005;46:963–71. [PubMed: 16108999]
152. Levy SE, Hyman SL. Complementary and alternative medicine treatments for children with autism spectrum disorders. Child Adolesc Psychiatr Clin N Am. 2008;17:803–20. [PubMed: 18775371]
153. Li H, Li Y, Shao J, Li R, Qin Y, Xie C, Zhao Z. The association analysis of RELN and GRM8 genes with autistic spectrum disorder in Chinese Han population. Am J Med Genet B Neuropsychiatr Genet. 2008;147B:194–200. [PubMed: 17955477]
154. Li H, Yamagata T, Mori M, Momoi MY. Absence of causative mutations and presence of autism-related allele in FOXP2 in Japanese autistic patients. Brain Dev. 2005;27:207–10. [PubMed: 15737702]
155. Li J, Nguyen L, Gleason C, Lotspeich L, Spiker D, Risch N, Myers RM. Lack of evidence for an association between WNT2 and RELN polymorphisms and autism. Am J Med Genet B Neuropsychiatr Genet. 2004;126B:51–7. [PubMed: 15048648]
156. Li J, Tabor HK, Nguyen L, Gleason C, Lotspeich LJ, Spiker D, Risch N, Myers RM. Lack of association between HoxA1 and HoxB1 gene variants and autism in 110 multiplex families. Am J Med Genet. 2002;114:24–30. [PubMed: 11840501]
157. Linday LA, Tsiouris JA, Cohen IL, Shindledecker R, DeCresce R. Famotidine treatment of children with autistic spectrum disorders: pilot research using single subject research design. J Neural Transm. 2001;108:593–611. [PubMed: 11459079]
158. Lintas C, Persico AM. Autistic phenotypes and genetic testing: state-of-the-art for the clinical geneticist. J Med Genet. 2009;46:1–8. [PubMed: 18728070]
159. Lisé MF, El-Husseini A. The neuroligin and neurexin families: from structure to function at the synapse. Cell Mol Life Sci. 2006;63:1833–49. [PubMed: 16794786]
160. Loesch DZ, Bui QM, Dissanayake C, Clifford S, Gould E, Bulhak-Paterson D, Tassone F, Taylor AK, Hessl D, Hagerman R, Huggins RM. Molecular and cognitive predictors of the continuum of autistic behaviours in fragile X. Neurosci Biobehav Rev. 2007;31:315–26. [PubMed: 17097142]
161. Lord C, McGee JP, Committee on Educational Interventions for Children with Autism, National Research Council. Educating Children with Autism. 2001. Washington, DC: The National Academies Press. Available at www.nap.edu. Accessed 4-10-10.
162. Lord C, Rutter M, Goode S, Heemsbergen J, Jordan H, Mawhood L, Schopler E. Autism diagnostic observation schedule: a standardized observation of communicative and social behavior. J Autism Dev Disord. 1989;19:185–212. [PubMed: 2745388]
163. Lord C, Rutter M, Le Couteur A. Autism Diagnostic Interview-Revised: a revised version of a diagnostic interview for caregivers of individuals with possible pervasive developmental disorders. J Autism Dev Disord. 1994;24:659–85. [PubMed: 7814313]
164. Lord C, Shulman C, DiLavore P. Regression and word loss in autistic spectrum disorders. J Child Psychol Psychiatry. 2004;45:936–55. [PubMed: 15225337]
165. Lukusa T, Vermeesch JR, Holvoet M, Fryns JP, Devriendt K. Deletion 2q37.3 and autism: molecular cytogenetic mapping of the candidate region for autistic disorder. Genet Couns. 2004;15:293–301. [PubMed: 15517821]
166. Ma DQ, Whitehead PL, Menold MM, Martin ER, Ashley-Koch AE, Mei H, Ritchie MD, DeLong GR, Abramson RK, Wright HH, Cuccaro ML, Hussman JP, Gilbert JR, Pericak-Vance MA. Identification of significant association and gene-gene interaction of GABA receptor subunit genes in autism. Am J Hum Genet. 2005;77:377–88. [PubMed: 16080114]
167. Maestrini E, Lai C, Marlow A, Matthews N, Wallace S, Bailey A, Cook EH, Weeks DE, Monaco AP. Serotonin transporter (5-HTT) and gamma-aminobutyric acid receptor subunit beta3 (GABRB3) gene polymorphisms are not associated with autism in the IMGSA families. The International Molecular Genetic Study of Autism Consortium. Am J Med Genet. 1999;88:492–6. [PubMed: 10490705]
168. Manning MA, Cassidy SB, Clericuzio C, Cherry AM, Schwartz S, Hudgins L, Enns GM, Hoyme HE. Terminal 22q deletion syndrome: a newly recognized cause of speech and language disability in the autism spectrum. Pediatrics. 2004;114:451–7. [PubMed: 15286229]
169. Manzi B, Loizzo AL, Giana G, Curatolo P. Autism and metabolic diseases. J Child Neurol. 2008;23:307–14. [PubMed: 18079313]
170. Marshall CR, Noor A, Vincent JB, Lionel AC, Feuk L, Skaug J, Shago M, Moessner R, Pinto D, Ren Y, Thiruvahindrapduram B, Fiebig A, Schreiber S, Friedman J, Ketelaars CE, Vos YJ, Ficicioglu C, Kirkpatrick S, Nicolson R, Sloman L, Summers A, Gibbons CA, Teebi A, Chitayat D, Weksberg R, Thompson A, Vardy C, Crosbie V, Luscombe S, Baatjes R, Zwaigenbaum L, Roberts W, Fernandez B, Szatmari P, Scherer SW. Structural variation of chromosomes in autism spectrum disorder. Am J Hum Genet. 2008;82:477–88. [PubMed: 18252227]
171. Martin CL, Duvall JA, Ilkin Y, Simon JS, Arreaza MG, Wilkes K, Alvarez-Retuerto A, Whichello A, Powell CM, Rao K, Cook E, Geschwind DH. Cytogenetic and molecular characterization of A2BP1/FOX1 as a candidate gene for autism. Am J Med Genet B Neuropsychiatr Genet. 2007;144B:869–76. [PubMed: 17503474]
172. Martin ER, Menold MM, Wolpert CM, Bass MP, Donnelly SL, Ravan SA, Zimmerman A, Gilbert JR, Vance JM, Maddox LO, Wright HH, Abramson RK, DeLong GR, Cuccaro ML, Pericak-Vance MA. Analysis of linkage disequilibrium in gamma-aminobutyric acid receptor subunit genes in autistic disorder. Am J Med Genet. 2000;96:43–8. [PubMed: 10686550]
173. McCarthy SE, Makarov V, Kirov G, Addington AM, McClellan J, Yoon S, Perkins DO, Dickel DE, Kusenda M, Krastoshevsky O, Krause V, Kumar RA, Grozeva D, Malhotra D, Walsh T, Zackai EH, Kaplan P, Ganesh J, Krantz ID, Spinner NB, Roccanova P, Bhandari A, Pavon K, Lakshmi B, Leotta A, Kendall J, Lee YH, Vacic V, Gary S, Iakoucheva LM, Crow TJ, Christian SL, Lieberman JA, Stroup TS, Lehtimäki T, Puura K, Haldeman-Englert C, Pearl J, Goodell M, Willour VL, Derosse P, Steele J, Kassem L, Wolff J, Chitkara N, McMahon FJ, Malhotra AK, Potash JB, Schulze TG, Nöthen MM, Cichon S, Rietschel M, Leibenluft E, Kustanovich V, Lajonchere CM, Sutcliffe JS, Skuse D, Gill M, Gallagher L, Mendell NR. Wellcome Trust Case Control Consortium, Craddock N, Owen MJ, O'Donovan MC, Shaikh TH, Susser E, Delisi LE, Sullivan PF, Deutsch CK, Rapoport J, Levy DL, King MC, Sebat J. Microduplications of 16p11.2 are associated with schizophrenia. Nat Genet. 2009;41:1223–7. [PubMed: 19855392]
174. McCauley JL, Olson LM, Delahanty R, Amin T, Nurmi EL, Organ EL, Jacobs MM, Folstein SE, Haines JL, Sutcliffe JS. A linkage disequilibrium map of the 1-Mb 15q12 GABA(A) receptor subunit cluster and association to autism. Am J Med Genet B Neuropsychiatr Genet. 2004a;131B:51–9. [PubMed: 15389768]
175. McCauley JL, Olson LM, Dowd M, Amin T, Steele A, Blakely RD, Folstein SE, Haines JL, Sutcliffe JS. Linkage and association analysis at the serotonin transporter (SLC6A4) locus in a rigid-compulsive subset of autism. Am J Med Genet B Neuropsychiatr Genet. 2004b;127B:104–12. [PubMed: 15108191]
176. McCoy PA, Shao Y, Wolpert CM, Donnelly SL, Ashley-Koch A, Abel HL, Ravan SA, Abramson RK, Wright HH, DeLong GR, Cuccaro ML, Gilbert JR, Pericak-Vance MA. No association between the WNT2 gene and autistic disorder. Am J Med Genet. 2002;114:106–9. [PubMed: 11840514]
177. McDougle CJ, Scahill L, Aman MG, McCracken JT, Tierney E, Davies M, Arnold LE, Posey DJ, Martin A, Ghuman JK, Shah B, Chuang SZ, Swiezy NB, Gonzalez NM, Hollway J, Koenig K, McGough JJ, Ritz L, Vitiello B. Risperidone for the core symptom domains of autism: results from the study by the autism network of the research units on pediatric psychopharmacology. Am J Psychiatry. 2005;162:1142–8. [PubMed: 15930063]
178. McGee GG, Morrier MJ, Daly T. An incidental teaching approach to early intervention for toddlers with autism. J Assoc Pers Severe. 1999;24:133–46.
179. Menold MM, Shao Y, Wolpert CM, Donnelly SL, Raiford KL, Martin ER, Ravan SA, Abramson RK, Wright HH, DeLong GR, Cuccaro ML, Pericak-Vance MA, Gilbert JR. Association analysis of chromosome 15 gabaa receptor subunit genes in autistic disorder. J Neurogenet. 2001;15:245–59. [PubMed: 12092907]
180. Mesibov G, Howley M. Accessing the Curriculum for Pupils with Autistic Spectrum Disorders. London: David Fulton Publishers; 2003.
181. Mesibov G, Shea V, Schopler E. The TEACCH Approach to Autism Spectrum Disorders. New York: Springer; 2004.
182. Miles JH, Hadden L, Takahashi TN, Hillman RE. Head circumference is an independent clinical finding associated with autism. Am J Med Genet. 2000;95:339–50. [PubMed: 11186888]
183. Miles JH, Hillman RE. Value of a clinical morphology examination in autism. Am J Med Genet. 2000;91:245–53. [PubMed: 10766977]
184. Miles JH, Takahashi TN, Bagby S, Sahota PK, Vaslow DF, Wang CH, Hillman RE, Farmer JE. Essential versus complex autism: definition of fundamental prognostic subtypes. Am J Med Genet A. 2005;135:171–80. [PubMed: 15887228]
185. Miles JH, Takahashi TN, Haber A, Hadden L. Autism families with a high incidence of alcoholism. J Autism Dev Disord. 2003;33:403–15. [PubMed: 12959419]
186. Miles JH, Takahashi TN, Hillman RE, Martin KL (2004) Autism symptoms are less severe in girls with essential autism. Am J Hum Genet .
187. Miles JH, Takahashi TN, Hong J, Munden N, Flournoy N, Braddock SR, Martin RA, Bocian ME, Spence MA, Hillman RE, Farmer JE. Development and validation of a measure of dysmorphology: useful for autism subgroup classification. Am J Med Genet A. 2008;146A:1101–16. [PubMed: 18383511]
188. Miller DT, Shen Y, Weiss LA, Korn J, Anselm I, Bridgemohan C, Cox GF, Dickinson H, Gentile J, Harris DJ, Hegde V, Hundley R, Khwaja O, Kothare S, Luedke C, Nasir R, Poduri A, Prasad K, Raffalli P, Reinhard A, Smith SE, Sobeih MM, Soul JS, Stoler J, Takeoka M, Tan WH, Thakuria J, Wolff R, Yusupov R, Gusella JF, Daly MJ, Wu BL. Microdeletion/duplication at 15q13.2q13.3 among individuals with features of autism and other neuropsychiatric disorders. J Med Genet. 2009;46:242–8. [PubMed: 18805830]
189. Moessner R, Marshall CR, Sutcliffe JS, Skaug J, Pinto D, Vincent J, Zwaigenbaum L, Fernandez B, Roberts W, Szatmari P, Scherer SW. Contribution of SHANK3 mutations to autism spectrum disorder. Am J Hum Genet. 2007;81:1289–97. [PubMed: 17999366]
190. Monaco AP, Bailey AJ. Autism. The search for susceptibility genes. Lancet. 2001;358 Suppl:S3. [PubMed: 11784552]
191. Moretti P, Zoghbi HY. MeCP2 dysfunction in Rett syndrome and related disorders. Curr Opin Genet Dev. 2006;16:276–81. [PubMed: 16647848]
192. Morrow EM, Yoo SY, Flavell SW, Kim TK, Lin Y, Hill RS, Mukaddes NM, Balkhy S, Gascon G, Hashmi A, Al Saad S, Ware J, Joseph RM, Greenblatt R, Gleason D, Ertelt JA, Apse KA, Bodell A, Partlow JN, Barry B, Yao H, Markianos K, Ferland RJ, Greenberg ME, Walsh CA. Identifying autism loci and genes by tracing recent shared ancestry. Science. 2008;321:218–23. [PubMed: 18621663]
193. Muhle R, Trentacoste SV, Rapin I. The genetics of autism. Pediatrics. 2004;113:e472–e486. [PubMed: 15121991]
194. Murch SH, Anthony A, Casson DH, Malik M, Berelowitz M, Dhillon AP, Thomson MA, Valentine A, Davies SE, Walker-Smith JA. Retraction of an interpretation. Lancet. 2004;363:750. [PubMed: 15016483]
195. Myers SM, Johnson CP. Management of children with autism spectrum disorders. Pediatrics. 2007;120:1162–82. [PubMed: 17967921]
196. Myles BS, Simpson RL. Asperger Syndrome: A Guide for Educators and Parents. 2 ed. Austin, TX: PRO-ED, Inc; 2003.
197. Myles BS, Simpson RL. Asperger syndrome: an overview of characteristics. Focus on Autism and Other Developmental Disabilities. Available at foa.sagepub.com. 2002. Accessed 4-10-10.
198. Naqvi S, Cole T, Graham JM. Cole-Hughes macrocephaly syndrome and associated autistic manifestations. Am J Med Genet. 2000;94:149–52. [PubMed: 10982971]
199. Newmeyer A, deGrauw T, Clark J, Chuck G, Salomons G. Screening of male patients with autism spectrum disorder for creatine transporter deficiency. Neuropediatrics. 2007;38:310–2. [PubMed: 18461508]
200. Niklasson L, Rasmussen P, Oskarsdottir S, Gillberg C. Autism, ADHD, mental retardation and behavior problems in 100 individuals with 22q11 deletion syndrome. Res Dev Disabil. 2009;30:763–73. [PubMed: 19070990]
201. Noor A, Marshall CR, Scherer SW, Vincent JB. Mutations in patched domain containing 1 gene (PTCHD1) are associated with autism spectrum disorder. Philadelphia, PA.: American Society of Human Genetics 58th Annual Meeting, 2008.
202. Nordin V, Gillberg C. The long-term course of autistic disorders: update on follow-up studies. Acta Psychiatr Scand. 1998;97:99–108. [PubMed: 9517902]
203. Nurmi EL, Bradford Y, Chen Y, Hall J, Arnone B, Gardiner MB, Hutcheson HB, Gilbert JR, Pericak-Vance MA, Copeland-Yates SA, Michaelis RC, Wassink TH, Santangelo SL, Sheffield VC, Piven J, Folstein SE, Haines JL, Sutcliffe JS. Linkage disequilibrium at the Angelman syndrome gene UBE3A in autism families. Genomics. 2001;77:105–13. [PubMed: 11543639]
204. Nurmi EL, Dowd M, Tadevosyan-Leyfer O, Haines JL, Folstein SE, Sutcliffe JS. Exploratory subsetting of autism families based on savant skills improves evidence of genetic linkage to 15q11-q13. J Am Acad Child Adolesc Psychiatry. 2003;42:856–63. [PubMed: 12819446]
205. Odom SL, Brown WH, Frey T, Karasu N, Smith-Canter LL, Strain PS. Evidence-Based Practices for Young Children with Autism: Contributions for Single-Subject Design Research. Focus on Autism and Other Developmental Disabilities. 2003;18:166–75.
206. Offit PA. Autism's False Prophets: Bad Science, Risky Medicine and the Search for a Cure. New York: Columbia University Press, 2008.
207. Oliveira G, Diogo L, Grazina M, Garcia P, Ataide A, Marques C, Miguel T, Borges L, Vicente AM, Oliveira CR. Mitochondrial dysfunction in autism spectrum disorders: a population-based study. Dev Med Child Neurol. 2005;47:185–9. [PubMed: 15739723]
208. O'Roak BJ, State MW. Autism genetics: strategies, challenges, and opportunities. Autism Res. 2008;1:4–17. [PubMed: 19360646]
209. Ozonoff S, Williams BJ, Gale S, Miller JN. Autism and autistic behavior in Joubert syndrome. J Child Neurol. 1999;14:636–41. [PubMed: 10511335]
210. Page DT, Kuti OJ, Prestia C, Sur M. Haploinsufficiency for Pten and Serotonin transporter cooperatively influences brain size and social behavior. Proc Natl Acad Sci U S A. 2009;106:1989–94. [PubMed: 19208814]
211. Pagnamenta AT, Wing K, Akha ES, Knight SJ, Bolte S, Schmotzer G, Duketis E, Poustka F, Klauck SM, Poustka A, Ragoussis J, Bailey AJ, Monaco AP. A 15q13.3 microdeletion segregating with autism. Eur J Hum Genet. 2009;17:687–92. [PubMed: 19050728]
212. Parisi MA, Dinulos MB, Leppig KA, Sybert VP, Eng C, Hudgins L. The spectrum and evolution of phenotypic findings in PTEN mutation positive cases of Bannayan-Riley-Ruvalcaba syndrome. J Med Genet. 2001;38:52–8. [PubMed: 11332402]
213. Persico AM, D'Agruma L, Maiorano N, Totaro A, Militerni R, Bravaccio C, Wassink TH, Schneider C, Melmed R, Trillo S, Montecchi F, Palermo M, Pascucci T, Puglisi-Allegra S, Reichelt KL, Conciatori M, Marino R, Quattrocchi CC, Baldi A, Zelante L, Gasparini P, Keller F. Reelin gene alleles and haplotypes as a factor predisposing to autistic disorder. Mol Psychiatry. 2001;6:150–9. [PubMed: 11317216]
214. Peterson P, Carta JJ, Greenwood C. Teaching enhanced milieu language teaching skills to parents in multiple risk families. J Early Intervention. 2005;27:94–109.
215. Petit E, Herault J, Martineau J, Perrot A, Barthelemy C, Hameury L, Sauvage D, Lelord G, Muh JP. Association study with two markers of a human homeogene in infantile autism. J Med Genet. 1995;32:269–74. [PubMed: 7643354]
216. Pickles A, Starr E, Kazak S, Bolton P, Papanikolaou K, Bailey A, Goodman R, Rutter M. Variable expression of the autism broader phenotype: findings from extended pedigrees. J Child Psychol Psychiatry. 2000;41:491–502. [PubMed: 10836679]
217. Piven J, Palmer P. Psychiatric disorder and the broad autism phenotype: evidence from a family study of multiple-incidence autism families. Am J Psychiatry. 1999;156:557–63. [PubMed: 10200734]
218. Polimeni MA, Richdale AL, Francis AJ. A survey of sleep problems in autism, Asperger's disorder and typically developing children. J Intellect Disabil Res. 2005;49:260–8. [PubMed: 15816813]
219. Pons R, Andreu AL, Checcarelli N, Vila MR, Engelstad K, Sue CM, Shungu D, Haggerty R, de Vivo DC, DiMauro S. Mitochondrial DNA abnormalities and autistic spectrum disorders. J Pediatr. 2004;144:81–5. [PubMed: 14722523]
220. Posey DJ, Stigler KA, Erickson CA, McDougle CJ. Antipsychotics in the treatment of autism. J Clin Invest. 2008;118:6–14. [PubMed: 18172517]
221. Potocki L, Bi W, Treadwell-Deering D, Carvalho CM, Eifert A, Friedman EM, Glaze D, Krull K, Lee JA, Lewis RA, Mendoza-Londono R, Robbins-Furman P, Shaw C, Shi X, Weissenberger G, Withers M, Yatsenko SA, Zackai EH, Stankiewicz P, Lupski JR. Characterization of Potocki-Lupski syndrome (dup(17)(p11.2p11.2)) and delineation of a dosage-sensitive critical interval that can convey an autism phenotype. Am J Hum Genet. 2007;80:633–49. [PubMed: 17357070]
222. Prizant BM, Wetherby AM, Rydell PJ. Communication intevention issues for children with autism spectrum disorders. In: Wetherby AM, Prizant BM, eds. Autism Spectrum Disorders: A Transational Developmental Perspective. Vol 9. Baltimore: Paul H Brookes Publishing Co; 2000:193-224.
223. Ramoz N, Reichert JG, Smith CJ, Silverman JM, Bespalova IN, Davis KL, Buxbaum JD. Linkage and association of the mitochondrial aspartate/glutamate carrier SLC25A12 gene with autism. Am J Psychiatry. 2004;161:662–9. [PubMed: 15056512]
224. Reddy KS. Cytogenetic abnormalities and fragile-X syndrome in Autism Spectrum Disorder. BMC Med Genet. 2005;6:3. [PubMed: 15655077]
225. Ritvo ER, Jorde LB, Mason-Brothers A, Freeman BJ, Pingree C, Jones MB, McMahon WM, Petersen PB, Jenson WR, Mo A. The UCLA-University of Utah epidemiologic survey of autism: recurrence risk estimates and genetic counseling. Am J Psychiatry. 1989;146:1032–6. [PubMed: 2750975]
226. Robins DL, Fein D, Barton ML, Green JA. The Modified Checklist for Autism in Toddlers: an initial study investigating the early detection of autism and pervasive developmental disorders. J Autism Dev Disord. 2001;31:131–44. [PubMed: 11450812]
227. Romano V, Cali F, Mirisola M, Gambino G, D' Anna R, Di Rosa P, Seidita G, Chiavetta V, Aiello F, Canziani F, De Leo G, Ayala GF, Elia M. Lack of association of HOXA1 and HOXB1 mutations and autism in Sicilian (Italian) patients. Mol Psychiatry. 2003;8:716–7. [PubMed: 12888798]
228. Roohi J, Montagna C, Tegay DH, Palmer LE, DeVincent C, Pomeroy JC, Christian SL, Nowak N, Hatchwell E. Disruption of contactin 4 in three subjects with autism spectrum disorder. J Med Genet. 2009;46:176–82. [PubMed: 18349135]
229. Rosenfeld JA, Coppinger J, Bejjani BA, Girirajan S, Eichler EE, Shaffer LG, Ballif BC. Speech delays and behavioral problems are the predominant features in individuals with developmental delays and 16p11.2 microdeletions and microduplications. J Neurodev Disord. 2010;2:26–38.
230. Sahoo T, Peters SU, Madduri NS, Glaze DG, German JR, Bird LM, Barbieri-Welge R, Bichell TJ, Beaudet AL, Bacino CA. Microarray based comparative genomic hybridization testing in deletion bearing patients with Angelman syndrome: genotype-phenotype correlations. J Med Genet. 2006;43:512–6. [PubMed: 16183798]
231. Sakurai T, Ramoz N, Reichert JG, Corwin TE, Kryzak L, Smith CJ, Silverman JM, Hollander E, Buxbaum JD. Association analysis of the NrCAM gene in autism and in subsets of families with severe obsessive-compulsive or self-stimulatory behaviors. Psychiatr Genet. 2006;16:251–7. [PubMed: 17106428]
232. Samaco RC, Hogart A, LaSalle JM. Epigenetic overlap in autism-spectrum neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A and GABRB3. Hum Mol Genet. 2005;14:483–92. [PubMed: 15615769]
233. Samaco RC, Nagarajan RP, Braunschweig D, LaSalle JM. Multiple pathways regulate MeCP2 expression in normal brain development and exhibit defects in autism-spectrum disorders. Hum Mol Genet. 2004;13:629–39. [PubMed: 14734626]
234. Schaefer GB, Lutz RE. Diagnostic yield in the clinical genetic evaluation of autism spectrum disorders. Genet Med. 2006;8:549–56. [PubMed: 16980810]
235. Schaefer GB, Mendelsohn NJ. Clinical genetics evaluation in identifying the etiology of autism spectrum disorders. Genet Med. 2008;10:301–5. [PubMed: 18414214]
236. Schechter R, Grether JK. Continuing increases in autism reported to California's developmental services system: mercury in retrograde. Arch Gen Psychiatry. 2008;65:19–24. [PubMed: 18180424]
237. Schenck A, Bardoni B, Langmann C, Harden N, Mandel JL, Giangrande A. CYFIP/Sra-1 controls neuronal connectivity in Drosophila and links the Rac1 GTPase pathway to the fragile X protein. Neuron. 2003;38:887–98. [PubMed: 12818175]
238. Schopler E, Reichler RJ, Renner BR. The Childhood Autism Rating Scale (CARS) for Diagnostic Screening and Classification of Autism. New York: Irvington Publishers; 1986.
239. Scott FJ, Baron-Cohen S, Bolton P, Brayne C. The CAST (Childhood Asperger Syndrome Test): preliminary development of a UK screen for mainstream primary-school-age children. Autism. 2002;6:9–31. [PubMed: 11918111]
240. Sebat J, Lakshmi B, Malhotra D, Troge J, Lese-Martin C, Walsh T, Yamrom B, Yoon S, Krasnitz A, Kendall J, Leotta A, Pai D, Zhang R, Lee YH, Hicks J, Spence SJ, Lee AT, Puura K, Lehtimäki T, Ledbetter D, Gregersen PK, Bregman J, Sutcliffe JS, Jobanputra V, Chung W, Warburton D, King MC, Skuse D, Geschwind DH, Gilliam TC, Ye K, Wigler M. Strong association of de novo copy number mutations with autism. Science. 2007;316:445–9. [PubMed: 17363630]
241. Segurado R, Conroy J, Meally E, Fitzgerald M, Gill M, Gallagher L. Confirmation of association between autism and the mitochondrial aspartate/glutamate carrier SLC25A12 gene on chromosome 2q31. Am J Psychiatry. 2005;162:2182–4. [PubMed: 16263864]
242. Selkirk CG, McCarthy Veach P, Lian F, Schimmenti L, LeRoy BS. Parents' perceptions of autism spectrum disorder etiology and recurrence risk and effects of their perceptions on family planning: Recommendations for genetic counselors. J Genet Couns. 2009;18:507–19. [PubMed: 19488842]
243. Seltzer MM, Shattuck P, Abbeduto L, Greenberg JS. Trajectory of development in adolescents and adults with autism. Ment Retard Dev Disabil Res Rev. 2004;10:234–47. [PubMed: 15666341]
244. Serajee FJ, Zhong H, Mahbubul Huq AH. Association of Reelin gene polymorphisms with autism. Genomics. 2006;87:75–83. [PubMed: 16311013]
245. Simons Foundation Autism Research Initeative. SFARI Gene. New York: Simons Foundation. Available at sfari.org. 2009.
246. Sharp AJ, Mefford HC, Li K, Baker C, Skinner C, Stevenson RE, Schroer RJ, Novara F, De Gregori M, Ciccone R, Broomer A, Casuga I, Wang Y, Xiao C, Barbacioru C, Gimelli G, Bernardina BD, Torniero C, Giorda R, Regan R, Murday V, Mansour S, Fichera M, Castiglia L, Failla P, Ventura M, Jiang Z, Cooper GM, Knight SJ, Romano C, Zuffardi O, Chen C, Schwartz CE, Eichler EE. A recurrent 15q13.3 microdeletion syndrome associated with mental retardation and seizures. Nat Genet. 2008;40:322–8. [PubMed: 18278044]
247. Shattuck PT. Diagnostic substitution and changing autism prevalence. Pediatrics. 2006;117:1438–9. [PubMed: 16585346]
248. Shinawi M, Liu P, Kang SH, Shen J, Belmont JW, Scott DA, Probst FJ, Craigen WJ, Graham B, Pursley A, Clark G, Lee J, Proud M, Stocco A, Rodriguez D, Kozel B, Sparagana S, Roeder E, McGrew S, Kurczynski T, Allison L, Amato S, Savage S, Patel A, Stankiewicz P, Beaudet A, Cheung SW, Lupski JR. Recurrent reciprocal 16p11.2 rearrangements associated with global developmental delay, behavioral problems, dysmorphism, epilepsy, and abnormal head size. J Med Genet. 2010;47:332–41. [PubMed: 19914906]
249. Shinawi M, Patel A, Panichkul P, Zascavage R, Peters SU, Scaglia F. The Xp contiguous deletion syndrome and autism. Am J Med Genet A. 2009a;149A:1138–48. [PubMed: 19441126]
250. Shinawi M, Schaaf CP, Bhatt SS, Xia Z, Patel A, Cheung SW, Lanpher B, Nagl S, Herding HS, Nevinny-Stickel C, Immken LL, Patel GS, German JR, Beaudet AL, Stankiewicz P. A small recurrent deletion within 15q13.3 is associated with a range of neurodevelopmental phenotypes. Nat Genet. 2009b;41:1269–71. [PubMed: 19898479]
251. Sikora DM, Pettit-Kekel K, Penfield J, Merkens LS, Steiner RD. The near universal presence of autism spectrum disorders in children with Smith-Lemli-Opitz syndrome. Am J Med Genet A. 2006;140:1511–8. [PubMed: 16761297]
252. Simpson R, de Boer-Ott S, Griewold D, Myles B, Byrd S, Ganz J, Cook K, Otten K, Ben-Arieh J, Kline S, Adams L. Autism Spectrum Disorders: Interventions and Treatments for Children and Youth. Thousand Oaks, CA: Corwin Press; 2005.
253. Simpson RL. Evidence-based practices and students with autism spectrum disorders. Focus on Autism and Other Developmental Disabilities. Available at foa.sagepub.com. 2005. Accessed 4-10-10.
254. Skaar DA, Shao Y, Haines JL, Stenger JE, Jaworski J, Martin ER, DeLong GR, Moore JH, McCauley JL, Sutcliffe JS, Ashley-Koch AE, Cuccaro ML, Folstein SE, Gilbert JR, Pericak-Vance MA. Analysis of the RELN gene as a genetic risk factor for autism. Mol Psychiatry. 2005;10:563–71. [PubMed: 15558079]
255. Skuse DH. Imprinting, the X-chromosome, and the male brain: explaining sex differences in the liability to autism. Pediatr Res. 2000;47:9–16. [PubMed: 10625077]
256. Smith M, Spence MA, Flodman P. Nuclear and mitochondrial genome defects in autisms. Ann N Y Acad Sci. 2009;1151:102–32. [PubMed: 19154520]
257. Solomon M, Goodlin-Jones BL, Anders TF. A social adjustment enhancement intervention for high functioning autism, Asperger's syndrome, and pervasive developmental disorder NOS. J Autism Dev Disord. 2004;34:649–68. [PubMed: 15679185]
258. Spence SJ, Schneider MT. The role of epilepsy and epileptiform EEGs in autism spectrum disorders. Pediatr Res. 2009;65:599–606. [PubMed: 19454962]
259. Splawski I, Timothy KW, Sharpe LM, Decher N, Kumar P, Bloise R, Napolitano C, Schwartz PJ, Joseph RM, Condouris K, Tager-Flusberg H, Priori SG, Sanguinetti MC, Keating MT. Ca(V)1.2 calcium channel dysfunction causes a multisystem disorder including arrhythmia and autism. Cell. 2004;119:19–31. [PubMed: 15454078]
260. Splawski I, Yoo DS, Stotz SC, Cherry A, Clapham DE, Keating MT. CACNA1H mutations in autism spectrum disorders. J Biol Chem. 2006;281:22085–91. [PubMed: 16754686]
261. Stathis SL, Cowley DM, Broe D. Autism and adenylosuccinase deficiency. J Am Acad Child Adolesc Psychiatry. 2000;39:274–5. [PubMed: 10714045]
262. Stefanatos GA. Regression in autistic spectrum disorders. Neuropsychol Rev. 2008;18:305–19. [PubMed: 18956241]
263. Stevens MC, Fein DA, Dunn M, Allen D, Waterhouse LH, Feinstein C, Rapin I. Subgroups of children with autism by cluster analysis: a longitudinal examination. J Am Acad Child Adolesc Psychiatry. 2000;39:346–52. [PubMed: 10714055]
264. Stoelb M, Yarnal R, Miles JH, Takahashi TN, Farmer J, McCathren R. Predicting responsiveness to treatment of children with autism: a retrospective study of the importance of physical dysmorphology. In: Focus on Autism and Other Developmental Disabilities. Available at foa.sagepub.com. 2004. Accessed 4-10-10.
265. Strain PS, Hoyson M. The Need for Longitudinal, Intensive Social Skill Intervention: LEAP Follow-Up Outcomes for Children with Autism. Topics in early childhood special education. 2000;20:116–22.
266. Stromland K, Sjogreen L, Miller M, Gillberg C, Wentz E, Johansson M, Nylen O, Danielsson A, Jacobsson C, Andersson J, Fernell E. Mobius sequence--a Swedish multidiscipline study. Eur J Paediatr Neurol. 2002;6:35–45. [PubMed: 11993954]
267. Stromme P, Mangelsdorf ME, Scheffer IE, Gecz J. Infantile spasms, dystonia, and other X-linked phenotypes caused by mutations in Aristaless related homeobox gene, ARX. Brain Dev. 2002;24:266–8. [PubMed: 12142061]
268. Sutcliffe JS, Delahanty RJ, Prasad HC, McCauley JL, Han Q, Jiang L, Li C, Folstein SE, Blakely RD. Allelic heterogeneity at the serotonin transporter locus (SLC6A4) confers susceptibility to autism and rigid-compulsive behaviors. Am J Hum Genet. 2005;77:265–79. [PubMed: 15995945]
269. Sutcliffe JS. Genetics. Insights into the pathogenesis of autism. Science. 2008;321:208–9. [PubMed: 18621658]
270. Szatmari P, Paterson AD, Zwaigenbaum L, Roberts W, Brian J, Liu XQ, Vincent JB, Skaug JL, Thompson AP, Senman L, Feuk L, Qian C, Bryson SE, Jones MB, Marshall CR, Scherer SW, Vieland VJ, Bartlett C, Mangin LV, Goedken R, Segre A, Pericak-Vance MA, Cuccaro ML, Gilbert JR, Wright HH, Abramson RK, Betancur C, Bourgeron T, Gillberg C, Leboyer M, Buxbaum JD, Davis KL, Hollander E, Silverman JM, Hallmayer J, Lotspeich L, Sutcliffe JS, Haines JL, Folstein SE, Piven J, Wassink TH, Sheffield V, Geschwind DH, Bucan M, Brown WT, Cantor RM, Constantino JN, Gilliam TC, Herbert M, Lajonchere C, Ledbetter DH, Lese-Martin C, Miller J, Nelson S, Samango-Sprouse CA, Spence S, State M, Tanzi RE, Coon H, Dawson G, Devlin B, Estes A, Flodman P, Klei L, McMahon WM, Minshew N, Munson J, Korvatska E, Rodier PM, Schellenberg GD, Smith M, Spence MA, Stodgell C, Tepper PG, Wijsman EM, Yu CE, Roge B, Mantoulan C, Wittemeyer K, Poustka A, Felder B, Klauck SM, Schuster C, Poustka F, Bolte S, Feineis-Matthews S, Herbrecht E, Schmotzer G, Tsiantis J, Papanikolaou K, Maestrini E, Bacchelli E, Blasi F, Carone S, Toma C, van Engeland H, de Jonge M, Kemner C, Koop F, Langemeijer M, Hijmans C, Staal WG, Baird G, Bolton PF, Rutter ML, Weisblatt E, Green J, Aldred C, Wilkinson JA, Pickles A, Le Couteur A, Berney T, McConachie H, Bailey AJ, Francis K, Honeyman G, Hutchinson A, Parr JR, Wallace S, Monaco AP, Barnby G, Kobayashi K, Lamb JA, Sousa I, Sykes N, Cook EH, Guter SJ, Leventhal BL, Salt J, Lord C, Corsello C, Hus V, Weeks DE, Volkmar F, Tauber M, Fombonne E, Shih A, Meyer KJ. Mapping autism risk loci using genetic linkage and chromosomal rearrangements. Nat Genet. 2007;39:319–28. [PubMed: 17322880]
271. Takahashi TN, Farmer JE, Deidrick KK, Hsu BS, Miles JH, Maria BL. Joubert syndrome is not a cause of classical autism. Am J Med Genet A. 2005;132:347–51. [PubMed: 15633174]
272. Takahashi TN, Miles JH. Diagnostic yield of a medical evaluation of children with autism and pervasive developmental disorders. In preparation. 2009.
273. Talebizadeh Z, Bittel DC, Veatch OJ, Butler MG, Takahashi TN, Miles JH. Do known mutations in neuroligin genes (NLGN3 and NLGN4) cause autism? J Autism Dev Disord. 2004;34:735–6. [PubMed: 15679194]
274. Taylor B. Vaccines and the changing epidemiology of autism. Child Care Health Dev. 2006;32:511–9. [PubMed: 16919130]
275. Tierney E, Bukelis I, Thompson RE, Ahmed K, Aneja A, Kratz L, Kelley RI. Abnormalities of cholesterol metabolism in autism spectrum disorders. Am J Med Genet B Neuropsychiatr Genet. 2006;141B:666–8. [PubMed: 16874769]
276. Tierney E, Nwokoro NA, Porter FD, Freund LS, Ghuman JK, Kelley RI. Behavior phenotype in the RSH/Smith-Lemli-Opitz syndrome. Am J Med Genet. 2001;98:191–200. [PubMed: 11223857]
277. Tse J, Strulovitch J, Tagalakis V, Meng L, Fombonne E. Social skills training for adolescents with Asperger syndrome and high-functioning autism. J Autism Dev Disord. 2007;37:1960–8. [PubMed: 17216559]
278. Tuchman R, Rapin I. Epilepsy in autism. Lancet Neurol. 2002;1:352–8. [PubMed: 12849396]
279. Turner G, Partington M, Kerr B, Mangelsdorf M, Gecz J. Variable expression of mental retardation, autism, seizures, and dystonic hand movements in two families with an identical ARX gene mutation. Am J Med Genet. 2002;112:405–11. [PubMed: 12376946]
280. Van den Berghe G, Vincent MF, Jaeken J. Inborn errors of the purine nucleotide cycle: adenylosuccinase deficiency. J Inherit Metab Dis. 1997;20:193–202. [PubMed: 9211192]
281. Van der Aa N, Rooms L, Vandeweyer G. van den EJ, Reyniers E, Fichera M, Romano C, Delle CB, Mortier G, Menten B, Destree A, Maystadt I, Mannik K, Kurg A, Reimand T, McMullan D, Oley C, Brueton L, Bongers EM, van Bon BW, Pfund R, Jacquemont S, Ferrarini A, Martinet D, Schrander-Stumpel C, Stegmann AP, Frints SG, de Vries BB, Ceulemans B, Kooy RF. Fourteen new cases contribute to the characterization of the 7q11.23 microduplication syndrome. Eur J Med Genet. 2009;52:94–100. [PubMed: 19249392]
282. Varga EA, Pastore M, Prior T, Herman GE, McBride KL. The prevalence of PTEN mutations in a clinical pediatric cohort with autism spectrum disorders, developmental delay, and macrocephaly. Genet Med. 2009;11:111–7. [PubMed: 19265751]
283. Veenstra-VanderWeele J, Cook EH. Molecular genetics of autism spectrum disorder. Mol Psychiatry. 2004;9:819–32. [PubMed: 15197396]
284. Viding E, Blakemore SJ. Endophenotype approach to developmental psychopathology: implications for autism research. Behav Genet. 2007;37:51–60. [PubMed: 16988798]
285. Vincent JB, Kolozsvari D, Roberts WS, Bolton PF, Gurling HM, Scherer SW. Mutation screening of X-chromosomal neuroligin genes: no mutations in 196 autism probands. Am J Med Genet B Neuropsychiatr Genet. 2004;129B:82–4. [PubMed: 15274046]
286. Volkmar F, Chawarska K, Klin A. Autism in infancy and early childhood. Annu Rev Psychol. 2005;56:315–36. [PubMed: 15709938]
287. Vorstman JA, Staal WG, van Daalen E, van Engeland H, Hochstenbach PF, Franke L. Identification of novel autism candidate regions through analysis of reported cytogenetic abnormalities associated with autism. Mol Psychiatry. 2006;11(1):18–28.
288. Wakefield AJ, Murch SH, Anthony A, Linnell J, Casson DM, Malik M, Berelowitz M, Dhillon AP, Thomson MA, Harvey P, Valentine A, Davies SE, Walker-Smith JA. Ileal-lymphoid-nodular hyperplasia, non-specific colitis, and pervasive developmental disorder in children. Lancet. 1998;351:637–41. [PubMed: 9500320]
289. Wakefield AJ. MMR vaccination and autism. Lancet. 1999;354:949–50. [PubMed: 10489978]
290. Wassink TH, Piven J, Patil SR. Chromosomal abnormalities in a clinic sample of individuals with autistic disorder. Psychiatric Genetics. 2001;11:57–63. [PubMed: 11525418]
291. Wassink TH, Piven J, Vieland VJ, Pietila J, Goedken RJ, Folstein SE, Sheffield VC. Evaluation of FOXP2 as an autism susceptibility gene. Am J Med Genet. 2002;114:566–9. [PubMed: 12116195]
292. Wassink TH, Piven J, Vieland VJ, Pietila J, Goedken RJ, Folstein SE, Sheffield VC. Examination of AVPR1a as an autism susceptibility gene. Mol Psychiatry. 2004;9:968–72. [PubMed: 15098001]
293. Webb BJ, Miller SP, Pierce TB, Strawser S, Jones WP. Effects of social skill instruction for high-functioning adolescents with autism spectrum disorders. In: Focus on Autism and Other Developmental Disabilities. Available at foa.sagepub.com. 2004. Accessed 4-10-10.
294. Weiss LA, Shen Y, Korn JM, Arking DE, Miller DT, Fossdal R, Saemundsen E, Stefansson H, Ferreira MA, Green T, Platt OS, Ruderfer DM, Walsh CA, Altshuler D, Chakravarti A, Tanzi RE, Stefansson K, Santangelo SL, Gusella JF, Sklar P, Wu BL, Daly MJ. Association between microdeletion and microduplication at 16p11.2 and autism. N Engl J Med. 2008;358:667–75. [PubMed: 18184952]
295. Weissman JR, Kelley RI, Bauman ML, Cohen BH, Murray KF, Mitchell RL, Kern RL, Natowicz MR. Mitochondrial disease in autism spectrum disorder patients: a cohort analysis. PLoS ONE. 2008;3:e3815. [PubMed: 19043581]
296. Werner E, Dawson G. Validation of the phenomenon of autistic regression using home videotapes. Arch Gen Psychiatry. 2005;62:889–95. [PubMed: 16061766]
297. Wetherby AM, Prizant BM. Communication and Symbolic Behavior Scales Developmental ProfileTM (CSBS DPTM). First normed edition. Baltimore, MD: Paul H Brookes; 2002.
298. Wilcox JA, Tsuang MT, Schnurr T, Baida-Fragoso N. Case-control family study of lesser variant traits in autism. Neuropsychobiology. 2003;47:171–7. [PubMed: 12824738]
299. Wing L, Shah A. Catatonia in autistic spectrum disorders. Br J Psychiatry. 2000;176:357–62. [PubMed: 10827884]
300. Wolfberg PJ. Play and imagination in children with autism. New York: Teachers College Press, 1999.
301. Wu JY, Kuban KC, Allred E, Shapiro F, Darras BT. Association of Duchenne muscular dystrophy with autism spectrum disorder. J Child Neurol. 2005b;20:790–5. [PubMed: 16417872]
302. Wu S, Jia M, Ruan Y, Liu J, Guo Y, Shuang M, Gong X, Zhang Y, Yang X, Zhang D. Positive association of the oxytocin receptor gene (OXTR) with autism in the Chinese Han population. Biol Psychiatry. 2005a;58:74–7. [PubMed: 15992526]
303. Xu S, Han JC, Morales A, Menzie CM, Williams K, Fan YS. Characterization of 11p14-p12 deletion in WAGR syndrome by array CGH for identifying genes contributing to mental retardation and autism. Cytogenet Genome Res. 2008;122:181–7. [PubMed: 19096215]
304. Yan J, Oliveira G, Coutinho A, Yang C, Feng J, Katz C, Sram J, Bockholt A, Jones IR, Craddock N, Cook EH, Vicente A, Sommer SS. Analysis of the neuroligin 3 and 4 genes in autism and other neuropsychiatric patients. Mol Psychiatry. 2005;10:329–32. [PubMed: 15622415]
305. Yang P, Lung FW, Jong YJ, Hsieh HY, Liang CL, Juo SH. Association of the homeobox transcription factor gene ENGRAILED 2 with autistic disorder in Chinese children. Neuropsychobiology. 2008;57:3–8. [PubMed: 18424904]
306. Yirmiya N, Pilowsky T, Nemanov L, Arbelle S, Feinsilver T, Fried I, Ebstein RP. Evidence for an association with the serotonin transporter promoter region polymorphism and autism. Am J Med Genet. 2001;105:381–6. [PubMed: 11378854]
307. Yirmiya N, Shaked M. Psychiatric disorders in parents of children with autism: a meta-analysis. J Child Psychol Psychiatry. 2005;46:69–83. [PubMed: 15660645]
308. Ylisaukko-Oja T, Rehnstrom K, Auranen M, Vanhala R, Alen R, Kempas E, Ellonen P, Turunen JA, Makkonen I, Riikonen R, Nieminen-von Wendt T, von Wendt L, Peltonen L, Jarvela I. Analysis of four neuroligin genes as candidates for autism. Eur J Hum Genet. 2005;13:1285–92. [PubMed: 16077734]
309. Young DJ, Bebbington A, Anderson A, Ravine D, Ellaway C, Kulkarni A, de Klerk N, Kaufmann WE, Leonard H. The diagnosis of autism in a female: could it be Rett syndrome? Eur J Pediatr. 2008;167:661–9. [PubMed: 17684768]
310. Zafeiriou DI, Ververi A, Vargiami E. Childhood autism and associated comorbidities. Brain Dev. 2007;29:257–72. [PubMed: 17084999]
311. Zhang H, Liu X, Zhang C, Mundo E, Macciardi F, Grayson DR, Guidotti AR, Holden JJ. Reelin gene alleles and susceptibility to autism spectrum disorders. Mol Psychiatry. 2002;7:1012–7. [PubMed: 12399956]
312. Zhong H, Serajee FJ, Nabi R, Huq AH. No association between the EN2 gene and autistic disorder. J Med Genet. 2003;40:e42. [PubMed: 12676915]
313. Zori RT, Marsh DJ, Graham GE, Marliss EB, Eng C. Germline PTEN mutation in a family with Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome. Am J Med Genet. 1998;80:399–402. [PubMed: 9856571]

Suggested Reading

1. Attwood T. The Complete Guide to Asperger's Syndrome. London: Jessica Kingsley Publishers, Ltd; 2007.
2. Chawarska K, Klin A, Volkmar FR,eds. Autism Spectrum Disorders in Infants and Toddlers, Diagnosis, Assessment, and Treatment. New York: The Guilford Press; 2008.
3. Goldstein S, Naglieri JA, Ozonoff S, eds. Assessment of Autism Spectrum Disorders. New York: The Guilford Press; 2009.
4. Immunization Safety Review Committee, Stratton K, Gable A, Shetty P, McCormick M, eds. Immunization Safety Review: Measles-Mumps-Rubella Vaccine and Autism. Washington, DC: The National Academies Press. Available at books.nap.edu. 2010. Accessed 4-10-10.
5. McAfee J. Navigating the Social World: A Curriculum for Individuals with Asperger's Syndrome, High Functioning Autism and Related Disorders. Arlington, TX: Future Horizons; 2002.
6. National Institute of Mental Health. Autism Spectrum Disorders: Pervasive Developmental Disorders. Available in pdf. 2004. Accessed 4-10-10.

Source:NCBI Bookshelf and GeneReviews

Note: Click on the title of the post to read the original post