Abstract

The WWOX gene was initially discovered as a putative tumor suppressor. More recently, its association with multiple central nervous system (CNS) pathologies has been recognized. WWOX biallelic germline pathogenic variants have been implicated in spinocerebellar ataxia type 12 (SCAR12; MIM:614322) and in early infantile epileptic encephalopathy (EIEE28; MIM:616211). WWOX germline copy number variants have also been associated with autism spectrum disorder (ASD). All identified germline genomic variants lead to partial or complete loss of WWOX function. Importantly, large-scale genome-wide association studies have also identified WWOX as a risk gene for common neurodegenerative conditions such as Alzheimer’s disease (AD) and multiple sclerosis (MS). Thus, the spectrum of CNS disorders associated with WWOX is broad and heterogeneous, and there is little understanding of potential mechanisms at play. Exploration of gene expression databases indicates that WWOX expression is comparatively higher in the human cerebellar cortex than in other CNS structures. However, RNA in-situ hybridization data from the Allen Mouse Brain Atlas show that specific regions of the basolateral amygdala (BLA), the medial entorhinal cortex (EC), and deep layers of the isocortex can be singled out as brain regions with specific higher levels of Wwox expression. These observations are in close agreement with single-cell RNA-seq data which indicate that neurons from the medial entorhinal cortex, Layer 5 from the frontal cortex as well as GABAergic basket cells and granule cells from cerebellar cortex are the specific neuronal subtypes that display the highest Wwox expression levels. Importantly, the brain regions and cell types in which WWOX is most abundantly expressed, such as the EC and BLA, are intimately linked to pathologies and syndromic conditions in turn associated with this gene, such as epilepsy, intellectual disability, ASD, and AD. Higher Wwox expression in interneurons and granule cells from cerebellum points to a direct link to the described cerebellar ataxia in cases of WWOX loss of function. We now know that total or partial impairment of WWOX function results in a wide and heterogeneous variety of neurodegenerative conditions for which the specific molecular mechanisms remain to be deciphered. Nevertheless, these observations indicate an important functional role for WWOX in normal development and function of the CNS. Evidence also indicates that disruption of WWOX expression at the gene or protein level in CNS has significant deleterious consequences.

Highlights

  • The WW domain-containing oxidoreductase gene (WWOX), originally discovered by our laboratory, maps to the ch16q23.1-23.2 region and encodes a 414-amino acid protein composed of two WW domains in its N-terminus and a central short-chain dehydrogenase/reductase (SDR) domain [1]

  • In order to evaluate comparative temporospatial expression of WWOX in human central nervous system (CNS), we explored the Human Brain Transcriptome (HBT) dataset, which is based on Affymetrix GeneChip arrays [17]

  • WWOX intrinsic and evolutionary conserved genomic fragility might represent a necessary feature of this gene as part of physiologic genomic rearrangements found in brain cells and proposed to play a role in the generation of neuronal diversity via somatic genomic mosaicism [55]. These observations emphasize the notion that WWOX, because containing FRA16D, is a hotspot for common germline copy number variants (CNVs) and it is precisely this intrinsic fragility that is the cause for the frequent association of WWOX CNVs with neurological and developmental disorders as described here, i.e., WWOX-Related Early Infantile Epileptic Encephalopathy (WOREE), autism spectrum disorder (ASD), intellectual disability (ID), and possibly other conditions, such as attention-deficit hyperactivity disorder (ADHD)

Read more

Summary

Introduction

The WW domain-containing oxidoreductase gene (WWOX), originally discovered by our laboratory, maps to the ch16q23.1-23.2 region and encodes a 414-amino acid protein composed of two WW domains in its N-terminus and a central short-chain dehydrogenase/reductase (SDR) domain [1]. Somatic deletions and translocations affecting WWOX accompanied by loss of expression are frequent in multiple cancer types and associated with tumor progression, therapy resistance, and poor disease outcomes [3,4]. Abundant evidence from multiple studies has accumulated causally linking WWOX loss of function with various central nervous system (CNS) pathologies. As it will be discussed human WWOX germline pathogenic variants have been directly implicated in complex and heterogeneous neurological disorders [5,6,7,8,9,10,11]. We discuss key evidence supporting the aforementioned conclusions while summarizing information from various CNS expression datasets and discussing cellular pathways in which WWOX is known to play relevant roles that are potentially associated with the various neurodegenerative conditions

WWOX Expression in CNS
WWOX-Associated CNS Disorders
Evidence from Rodent Models of Wwox Ablation
WWOX as a Risk Locus for Alzheimer’s Disease
Findings
Conclusions
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