Chapter Five - MET Receptor Tyrosine Kinase as an Autism Genetic Risk Factor

https://doi.org/10.1016/B978-0-12-418700-9.00005-8Get rights and content

Abstract

In this chapter, we will briefly discuss recent literature on the role of MET receptor tyrosine kinase (RTK) in brain development and how perturbation of MET signaling may alter normal neurodevelopmental outcomes. Recent human genetic studies have established MET as a risk factor for autism, and the molecular and cellular underpinnings of this genetic risk are only beginning to emerge from obscurity. Unlike many autism risk genes that encode synaptic proteins, the spatial and temporal expression pattern of MET RTK indicates this signaling system is ideally situated to regulate neuronal growth, functional maturation, and establishment of functional brain circuits, particularly in those brain structures involved in higher levels of cognition, social skills, and executive functions.

Introduction

Autism spectrum disorders (ASD), which include autistic disorder, Asperger's syndrome, and pervasive developmental disorder (PDD)-not otherwise specified, are a group of neurodevelopmental syndromes that share a disease onset during early brain development and maturation (Abrahams and Geschwind, 2008, Geschwind and Levitt, 2007, Walsh et al., 2008). There have been no unifying neuropathologic or neurobiological features that define ASDs. The diagnosis is based on clinical assessment of some core behavioral features, including impaired communicative skills, atypical social behavior, and restricted interests and repetitive behaviors. Two cardinal features of ASD are heritability and heterogeneity. Heritability refers to the fact that autism has evidently the strongest genetic components of all the developmental neuropsychiatric disorders. This is exemplified by the 82–92% concordance rate for autism among monozygotic twins as compared with ~ 10% concordance rate for dizygotic twins (Abrahams and Geschwind, 2008, Bailey et al., 1995, Constantino et al., 2013). Heterogeneity is reflected by the enormous number (> 200) of gene loci (Aldinger et al., 2011, Ebert and Greenberg, 2013, Piggot et al., 2009) that contribute to the risk of developing ASD, hence imposing a major challenge for the identification of causative genes. While this genetic heterogeneity can manifest as noncoding variations, de novo mutations that produce syndromic disorders with autistic traits, copy number variations, and chromosome abnormalities (Marshall et al., 2008, Nakatani et al., 2009, Piggot et al., 2009, Sebat et al., 2007, Walsh et al., 2008), their functional implication spans even wider, from neuronal growth, projection and motility, GTPase/Ras-mediated signaling and cytoskeletal organization, proteolysis, to activity-dependent synaptic remodeling (Levitt and Campbell, 2009, Pinto et al., 2010). Thus, to gain insights into the underlying mechanisms of ASD will require a multidisciplinary approach focusing on brain regions, neural networks, and cellular substrates.

ASD is a complex disorder and, as such, identification of causative genes has been hampered by many inherent problems, such as multiple gene effects/interactions, environmental factors, gene–environment interactions, variable penetrance for each individual gene, and genetic heterogeneity. Many well-established autism risk genes encode proteins that are involved in the molecular networks controlling formation and function of the glutamatergic synapse, the submicron-scale structure that connects individual neurons into functional networks capable of computational outputs. These well-established genes include, but are not limited to, NRXN1, PTEN, SHANK3, UBE3a, NF1, NLGN3/4, CNTNAP2, SYNGAP1, and FMR1 (Alarcon et al., 2008, Bourgeron, 2009, Clement et al., 2012, Durand et al., 2007, Penagarikano et al., 2011, Piggot et al., 2009, Tabuchi et al., 2007, Yashiro et al., 2009). These molecules function by mediating pre- and postsynaptic assembly, scaffolding the synaptic structure, controlling neurotransmitter release, and affecting the activity-dependent structural changes, processes critical to sculpting our experience into neuronal circuits to guide future behavior. Not surprisingly, pathogenic mutations of the previously mentioned ASD genes during development have been shown to lead to synaptic dysfunction, impact the brain circuit, and disrupt the balanced excitatory/inhibitory brain networks (Ebert and Greenberg, 2013, Rubenstein and Merzenich, 2003, Tabuchi et al., 2007).

It is important to note, however, that synaptogenesis and neural circuit dynamics are relatively late events during the neurodevelopmental timeline. Prior to these events, the production and positioning of neurons in a correct cellular and network context must take place in order for synaptogenesis and circuit remodeling to occur. These early histogenic events are determined by genetic programs encoding neurogenesis, migration, neurite outgrowth and polarization, and axon guidance at critical developmental stages. At the cellular level, once a neuron is born, it migrates a long distance before arriving at its destination and differentiating. Neurons extend two classes of processes: a single axon to carry its output and several dendrites to collect information input. Once this neuronal polarity is established, the axon navigates through a complex environment to find its target, and dendrites undergo extensive growth and branching. The last step in forming functional circuitry is the establishment of synaptic connections between different neurons (Bradke and Dotti, 2000, Craig and Banker, 1994, Mueller, 1999, Tessier-Lavigne and Goodman, 1996). Two major types of synapses, excitatory and inhibitory, coexist within any functional circuitry, and their balanced action on the postsynaptic neurons shapes their functional output (Rubenstein & Merzenich, 2003). Therefore, aberrant genetic programs during this early extended timeline (as compared to impaired synaptic function at later stages) may profoundly affect brain function as well. Consistently, autism risk genes have been shown to control wide aspects of developmental events including neurogenesis, synaptogenesis, glutamatergic transmission, endosomal trafficking, and protein turnover (Ebert and Greenberg, 2013, Qiu et al., 2012, Walsh et al., 2008). As diverse as these risk genes appear, they may converge on a final common molecular pathway to disrupt developmental outcomes that perturb circuit formation and maturation.

The development of the central nervous system (CNS) is a complex process driven by a myriad of factors including a large family of growth factors and their receptors. Protein receptor tyrosine kinases (RTKs), which are cell-surface receptors for many polypeptide growth factors, hormones, and cytokines (Robinson, Wu, & Lin, 2000), regulate many aspects of neuronal physiology, including neurogenesis and survival, differentiation and migration, patterned connectivity, and plasticity. The human gene MET, which encodes MET RTK (Cooper et al., 1984), has emerged as a prominent risk factor for ASD (Campbell et al., 2006, Campbell et al., 2009, Jackson et al., 2009, Sousa et al., 2009, Thanseem et al., 2010). MET plays a pleiotropic role in cell proliferation, motogenesis, differentiation, and survival in many tissue types (Birchmeier et al., 2003, Maina et al., 1998). The ligand for MET receptor, hepatocyte growth factor (HGF), is a polypeptide growth factor that activates MET (Naldini, Weidner, et al., 1991). Both MET and HGF are expressed in the developing brain, with distinct spatial and temporal profiles (Judson, Amaral and Levitt, 2011, Judson et al., 2009, Jung et al., 1994). Genetic studies from multiple laboratories have found that functional MET promoter variants are associated with differential risks for ASD. Consistently, clinical imaging and animal studies have provided evidence that disrupted MET signaling levels produce both morphological and functional alterations in neurons in those brain regions implicated in producing the ASD endophenotypes. In this chapter, we will briefly discuss how MET signaling might be ideally situated to regulate circuits and modify neuronal function. We review recent literature and hypothesize that MET signaling plays a critical role in balancing neuronal growth, functional maturation, and establishing functional circuits.

Section snippets

MET Receptor Tyrosine Kinase-Mediated Signaling has a Pleiotropic Role in Multiple Organ Ontogenesis

The MET RTK and its sole polypeptide growth factor ligand, HGF, exemplify a versatile signaling system that has effects not only on neurons but also on multiple target tissues during embryogenesis. HGF, also known as “scatter factor,” was originally identified as a molecule capable of triggering proliferation, motility, and morphogenesis in many epithelial cell types and is also involved in organ regeneration, angiogenesis, and tumor invasion (Naldini, Vigna, et al., 1991). The MET receptor was

MET Signaling Plays a Role in a Large Number of Neurodevelopment Events

The molecular signaling events discussed earlier are mostly ascertained in human epithelial or cancer cell lines, and, collectively, they mediate cell growth and invasive programs. The recognition of MET serving as a key signaling component in specific neurodevelopmental events is relatively new compared with the well-established roles in cancer biology (Judson, Eagleson and Levitt, 2011, Maina et al., 1998). It is currently unclear to what extent these signaling events are operating in neurons

MET Receptor Tyrosine Kinase Expression in the Developing Brain

To better understand the developmental capacity of MET/HGF signaling in the brain, it is important to ascertain the normal spatiotemporal patterns of MET/HGF expression levels. Several early studies have attempted to resolve this question. Di Renzo et al. (1993) showed that MET is expressed in the human CNS and MET protein is detectable in human brain tissues using Western blot. Immunohistochemical staining of MET revealed a rather extensive labeling of both gray and white matter, particularly

The Human MET Gene Emerges as a Prominent Autism Risk Factor

The human MET gene (OMIM 164860; chromosome 7q31) was first reported by Campbell et al. as a risk factor for autism based on genome-wide association studies aimed to identify genetic variants that are overrepresented in individuals with autism compared to control populations (Campbell et al., 2006). MET was hypothesized as a candidate gene based on the following observations prior to this study: First, MET is located on human chromosome 7q31, under a linkage peak identified in multiple

Implication of MET Signaling in Neural Development and Functional Connectivity

ASD is considered a developmental disconnection syndrome, and the core pathophysiological basis can likely be attributed to disrupted ontogeny of neural connectivity (Courchesne and Pierce, 2005, Geschwind and Levitt, 2007). The specificity and the timing of brain circuits that are involved and the severity of disruption determine the presentation of clinical phenotypes. There is strong molecular and cellular basis for this hypothesized miswiring. MET signaling is required for multiple

Concluding Remarks

Translating the genetic contributions to neurodevelopmental disorders, such as ASD, into pathophysiological mechanisms will bridge the current knowledge gap and facilitate developing novel interventions and treatments. Many of the most compelling candidate genes identified for rare/syndromic and idiopathic forms of ASD so far are involved in brain wiring and synaptic function by being an integral part of synaptic molecular machinery, by regulating gene transcriptions, or by contributing to the

Acknowledgments

The authors thank Dr. Aaron McGee, Zhongming Lu, and Mariel Piechowicz for proofreading and their critiques of this chapter.

References (128)

  • F. Davey et al.

    Cooperation between HGF and CNTF in promoting the survival and growth of sensory and parasympathetic neurons

    Molecular and Cellular Neurosciences

    (2000)
  • A. Ebens et al.

    Hepatocyte growth factor/scatter factor is an axonal chemoattractant and a neurotrophic factor for spinal motor neurons

    Neuron

    (1996)
  • D.H. Geschwind et al.

    Autism spectrum disorders: Developmental disconnection syndromes

    Current Opinion in Neurobiology

    (2007)
  • S. Hong et al.

    Detecting abnormalities of corpus callosum connectivity in autism using magnetic resonance imaging and diffusion tensor tractography

    Psychiatry Research

    (2011)
  • L.H. Kawas et al.

    Nanoscale mapping of the Met receptor on hippocampal neurons by AFM and confocal microscopy

    Nanomedicine

    (2013)
  • C.S. Lim et al.

    Hepatocyte growth factor and c-Met promote dendritic maturation during hippocampal neuron differentiation via the Akt pathway

    Cellular Signalling

    (2008)
  • L. Luo et al.

    Genetic dissection of neural circuits

    Neuron

    (2008)
  • F. Maina et al.

    Uncoupling of Grb2 from the Met receptor in vivo reveals complex roles in muscle development

    Cell

    (1996)
  • F. Maina et al.

    Multiple roles for hepatocyte growth factor in sympathetic neuron development

    Neuron

    (1998)
  • F. Maina et al.

    Coupling Met to specific pathways results in distinct developmental outcomes

    Molecular Cell

    (2001)
  • C.J. Marshall

    Specificity of receptor tyrosine kinase signaling: Transient versus sustained extracellular signal-regulated kinase activation

    Cell

    (1995)
  • C.R. Marshall et al.

    Structural variation of chromosomes in autism spectrum disorder

    American Journal of Human Genetics

    (2008)
  • J. Nakatani et al.

    Abnormal behavior in a chromosome-engineered mouse model for human 15q11-13 duplication seen in autism

    Cell

    (2009)
  • L.E. O'Brien et al.

    ERK and MMPs sequentially regulate distinct stages of epithelial tubule development

    Developmental Cell

    (2004)
  • O. Penagarikano et al.

    Absence of CNTNAP2 leads to epilepsy, neuronal migration abnormalities, and core autism-related deficits

    Cell

    (2011)
  • J. Piggot et al.

    Neural systems approaches to the neurogenetics of autism spectrum disorders

    Neuroscience

    (2009)
  • C. Ponzetto et al.

    A multifunctional docking site mediates signaling and transformation by the hepatocyte growth factor/scatter factor receptor family

    Cell

    (1994)
  • C. Ponzetto et al.

    Specific uncoupling of GRB2 from the Met receptor. Differential effects on transformation and motility

    The Journal of Biological Chemistry

    (1996)
  • E.M. Powell et al.

    Hepatocyte growth factor/scatter factor is a motogen for interneurons migrating from the ventral to dorsal telencephalon

    Neuron

    (2001)
  • K. Pozo et al.

    Unraveling mechanisms of homeostatic synaptic plasticity

    Neuron

    (2010)
  • J.D. Rudie et al.

    Autism-associated promoter variant in MET impacts functional and structural brain networks

    Neuron

    (2012)
  • C.P. Sahyoun et al.

    Neuroimaging of the functional and structural networks underlying visuospatial vs. linguistic reasoning in high-functioning autism

    Neuropsychologia

    (2010)
  • B.S. Abrahams et al.

    Advances in autism genetics: On the threshold of a new neurobiology

    Nature Reviews Genetics

    (2008)
  • J.S. Anderson et al.

    Decreased interhemispheric functional connectivity in autism

    Cerebral Cortex

    (2011)
  • L. Bai et al.

    Hepatocyte growth factor mediates mesenchymal stem cell-induced recovery in multiple sclerosis models

    Nature Neuroscience

    (2012)
  • A. Bailey et al.

    Autism as a strongly genetic disorder: Evidence from a British twin study

    Psychological Medicine

    (1995)
  • C. Birchmeier et al.

    Met, metastasis, motility and more

    Nature Reviews Molecular Cell Biology

    (2003)
  • F. Bladt et al.

    Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud

    Nature

    (1995)
  • C. Boccaccio et al.

    Induction of epithelial tubules by growth factor HGF depends on the STAT pathway

    Nature

    (1998)
  • D.P. Bottaro et al.

    Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product

    Science

    (1991)
  • D.B. Campbell et al.

    Distinct genetic risk based on association of MET in families with co-occurring autism and gastrointestinal conditions

    Pediatrics

    (2009)
  • D.B. Campbell et al.

    Disruption of cerebral cortex MET signaling in autism spectrum disorder

    Annals of Neurology

    (2007)
  • D.B. Campbell et al.

    Genetic evidence implicating multiple genes in the MET receptor tyrosine kinase pathway in autism spectrum disorder

    Autism Research

    (2008)
  • D.B. Campbell et al.

    A genetic variant that disrupts MET transcription is associated with autism

    Proceedings of the National Academy of Sciences of the United States of America

    (2006)
  • A. Caton et al.

    The branchial arches and HGF are growth-promoting and chemoattractant for cranial motor axons

    Development

    (2000)
  • J.N. Constantino et al.

    Autism recurrence in half siblings: Strong support for genetic mechanisms of transmission in ASD

    Molecular Psychiatry

    (2013)
  • C.S. Cooper et al.

    Molecular cloning of a new transforming gene from a chemically transformed human cell line

    Nature

    (1984)
  • A.M. Craig et al.

    Neuronal polarity

    Annual Review of Neuroscience

    (1994)
  • M.F. Di Renzo et al.

    Selective expression of the Met/HGF receptor in human central nervous system microglia

    Oncogene

    (1993)
  • C.M. Durand et al.

    Mutations in the gene encoding the synaptic scaffolding protein SHANK3 are associated with autism spectrum disorders

    Nature Genetics

    (2007)
  • Cited by (37)

    • Receptor tyrosine kinases (RTKs): from biology to pathophysiology

      2023, Receptor Tyrosine Kinases in Neurodegenerative and Psychiatric Disorders
    • Neurodevelopmental disorders, immunity, and cancer are connected

      2022, iScience
      Citation Excerpt :

      Stimulated receptor tyrosine kinases (RTKs), a major activating arm of MAPK, toll-like receptors (TLRs), and interleukin receptors (IL-Rs), which activate MAPKs in immune cells (discussed below in the review), transduce signals through active mediators, such as growth factor bound protein 2 (Grb2) and SHP2 (SH2 domain-containing phosphatase 2). The RTKs, e.g., Met receptor tyrosine kinase in autism spectrum disorder (ASD), ERBB4 (erb-b2 receptor tyrosine kinase 4) in ASD and schizophrenia susceptibility (Judson et al., 2011; Norton et al., 2006), and their mediators are also players in neurodevelopmental disorders (Ma and Qiu, 2020; Peng et al., 2013; Tomita et al., 2020), immunity (Crawley et al., 2016), and inflammation (Kaminska, 2005). Proliferation also requires cell growth, a role which can be fulfilled by major pathways such as Ras/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) (Borrie et al., 2017; Papa and Pandolfi, 2019; Wang et al., 2017; White et al., 2020), which like MAPK also feed into the cell cycle at the G1 phase (Nussinov et al., 2016b).

    • A Short Isoform of Coagulation Factor XII mRNA Is Expressed by Neurons in the Human Brain

      2019, Neuroscience
      Citation Excerpt :

      Further investigation into the function of the short FXII isoform may shed light on its role in these cell populations. In addition to the insights on the role of neuronal HGF-Met signaling derived from in vitro and animal studies, accumulating evidence suggests that dysregulation in HGF-Met signaling contributes to neurodevelopmental disorders (Peng et al., 2013; Eagleson et al., 2017) and neurodegeneration (Wright and Harding, 2015) in humans. In particular, expression of Met protein is decreased in the hippocampal neurons of AD patients (Hamasaki et al., 2014), while HGF protein levels are increased in AD brain (Fenton et al., 1998).

    • Immune Dysfunction in Autism Spectrum Disorder

      2016, Neuronal and Synaptic Dysfunction in Autism Spectrum Disorder and Intellectual Disability
    View all citing articles on Scopus
    View full text