Abstract

Autism spectrum disorder (ASD) has captured the attention of scientists, clinicians and the lay public because of its uncertain origins and striking and unexplained clinical heterogeneity. Here we review genetic, genomic, cellular, postmortem, animal model, and cell model evidence that shows ASD begins in the womb. This evidence leads to a new theory that ASD is a multistage, progressive disorder of brain development, spanning nearly all of prenatal life. ASD can begin as early as the 1st and 2nd trimester with disruption of cell proliferation and differentiation. It continues with disruption of neural migration, laminar disorganization, altered neuron maturation and neurite outgrowth, disruption of synaptogenesis and reduced neural network functioning. Among the most commonly reported high-confidence ASD (hcASD) genes, 94% express during prenatal life and affect these fetal processes in neocortex, amygdala, hippocampus, striatum and cerebellum. A majority of hcASD genes are pleiotropic, and affect proliferation/differentiation and/or synapse development. Proliferation and subsequent fetal stages can also be disrupted by maternal immune activation in the 1st trimester. Commonly implicated pathways, PI3K/AKT and RAS/ERK, are also pleiotropic and affect multiple fetal processes from proliferation through synapse and neural functional development. In different ASD individuals, variation in how and when these pleiotropic pathways are dysregulated, will lead to different, even opposing effects, producing prenatal as well as later neural and clinical heterogeneity. Thus, the pathogenesis of ASD is not set at one point in time and does not reside in one process, but rather is a cascade of prenatal pathogenic processes in the vast majority of ASD toddlers. Despite this new knowledge and theory that ASD biology begins in the womb, current research methods have not provided individualized information: What are the fetal processes and early-age molecular and cellular differences that underlie ASD in each individual child? Without such individualized knowledge, rapid advances in biological-based diagnostic, prognostic, and precision medicine treatments cannot occur. Missing, therefore, is what we call ASD Living Biology. This is a conceptual and paradigm shift towards a focus on the abnormal prenatal processes underlying ASD within each living individual. The concept emphasizes the specific need for foundational knowledge of a living child’s development from abnormal prenatal beginnings to early clinical stages. The ASD Living Biology paradigm seeks this knowledge by linking genetic and in vitro prenatal molecular, cellular and neural measurements with in vivo post-natal molecular, neural and clinical presentation and progression in each ASD child. We review the first such study, which confirms the multistage fetal nature of ASD and provides the first in vitro fetal-stage explanation for in vivo early brain overgrowth. Within-child ASD Living Biology is a novel research concept we coin here that advocates the integration of in vitro prenatal and in vivo early post-natal information to generate individualized and group-level explanations, clinically useful prognoses, and precision medicine approaches that are truly beneficial for the individual infant and toddler with ASD.

Highlights

  • Electronic supplementary material The online version of this article contains supplementary material, which is available to authorized users.Extended author information available on the last page of the articleAutism spectrum disorder (ASD) has captured the attention of scientists and clinicians, as well as the lay public, in part because the clinical profile is so striking [1,2,3]

  • There is a need to bridge the gap between fetal and postnatal biology for living ASD toddlers, and this requires a paradigm shift. This paradigm we describe here, ASD Living Biology, approaches the fundamental questions above, by acquiring fetal and early post-natal measures using iPS cells derived from skin or blood at any age, and integrating them using a within-subjects design

  • An excess or reduction of synapse formation may occur in different ASD cases; excess brain growth occurs in most while reduced size occurs in a small subset

Read more

Summary

Introduction

The hcASD genes SHANK3, FMR1, CTNND2, CHD8, SYNGAP1, MECP2, PTEN, ARID1B and NF1 are associated with dysregulation of neurite outgrowth and neuronal function through the PI3K/AKT, RAS/ERK, WNT and βcatenin signaling pathways (Table 1) These pathways control soma size, dendritic arborization, axon generation, spine development and synapse function [184,185,186,187,188]. Upregulated cell cycle and downregulated migration and neurite outgrowth gene expression Cortical layering: over-production of neurons, increased cortical thickness, focal cortical dysplasia Cerebellar vermis dysplasia Microglia: enhanced priming, activation Dendritic morphology abnormalities GABAergic signaling, excitatory/inhibitory imbalance, number of interneurons White matter neuron density Dendritic spines number and turnover rates Synaptic pruning and proteins Early brain overgrowth Myelin functionality and stability Dopamine system Serotonin levels ASD-like abnormal social, vocalization, and ritualistic behaviors Gender-dependent effects Transgenerational effects. The more atypical such post-natal experience becomes, the more an individual’s development could become canalized for atypical outcomes

Conclusions
Findings
Compliance with ethical standards
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call