WWOX (WW domain-containing oxidoreductase) is a tumor suppressor gene located on chromosome 16q23.3–24.1, spanning 1.1 million bases within the fragile site FRA16D . It encodes a 414-amino-acid protein (46 kDa) with two N-terminal WW domains for protein-protein interactions and a C-terminal short-chain dehydrogenase/reductase (SDR) domain linked to metabolic regulation . WWOX is ubiquitously expressed but shows high activity in hormone-regulated tissues (e.g., breast, prostate) and the central nervous system (CNS) .
Tumor Suppression: Loss of heterozygosity (LOH) in 70% of breast cancers and glioblastomas; regulates apoptosis via interactions with p53 and JNK pathways .
Neurodevelopment: Critical for neuronal differentiation, myelination, and synaptic plasticity; mutations linked to epileptic encephalopathy (WOREE) and spinocerebellar ataxia (SCAR12) .
Metabolic Regulation: Modulates glycolysis, lipid metabolism, and steroidogenesis; deficiency disrupts glucose/HDL homeostasis .
Apoptosis Regulation:
Metabolic Dysregulation:
3D Growth Defects:
MGSSHHHHHH SSGLVPRGSH MAALRYAGLD DTDSEDELPP GWEERTTKDG WVYYANHTEE KTQWEHPKTG KRKRVAGDLP YGWEQETDEN GQVFFVDHIN KRTTYLDPRL AFTVDDNPTK PTTRQRYDGS TTAMEILQGR DFTGKVVVVT GANSGIGFET AKSFALHGAH VILACRNMAR ASEAVSRILE EWQQGAATTV YCAAVPELEG LGGMYFNNCC RCMPSPEAQS EETARTLWAL SERLIQERLG SQSG.
The WWOX (WW domain-containing oxidoreductase) gene is located in the chromosomal common fragile site FRA16D and encodes a tumor suppressor protein containing WW domains and an oxidoreductase domain. It plays critical roles in maintaining central nervous system homeostasis and contributes to genomic stability . The WWOX protein is highly conserved evolutionarily, with human WWOX showing 93% identity and 95% similarity to mouse Wwox at the protein sequence level . The gene functions in multiple cellular processes including regulation of neuronal differentiation and migration in the brain, with its dysfunction leading to serious neurodevelopmental disorders in humans .
Germline biallelic mutations in the WWOX gene lead to two major clinical phenotypes:
WOREE syndrome (WWOX-related epileptic encephalopathy, also known as developmental and epileptic encephalopathy 28 or DEE28, OMIM 616211): Characterized by early-onset severe epilepsy with variable seizure manifestations (tonic, clonic, tonic-clonic, myoclonic, infantile spasms, and absence seizures), severe developmental delay, and profound disability. Most affected patients cannot make eye contact, sit, speak, or walk .
SCAR12 (spinocerebellar ataxia, autosomal recessive 12, OMIM 614322): A milder phenotype typically caused by missense mutations in WWOX, featuring ataxia and epilepsy that may be responsive to antiepileptic drugs, though patients still display ataxia and intellectual disability .
Additionally, WWOX mutations have been documented in patients with West syndrome, characterized by epileptic spasms with hypsarrhythmia .
Several animal models have been developed to study WWOX function and dysfunction:
Wwox-knockout mice: Complete ablation of Wwox leads to postnatal lethality and recapitulates many of the severe phenotypes seen in WOREE syndrome, including intractable epilepsy, hypomyelination, and early death . These models demonstrate that targeted disruption of the Wwox gene causes neurodevelopmental disorders including abnormal neuronal differentiation and migration in the brain, cerebral malformations such as microcephaly, neuronal disorganization, and defective cerebellar midline fusion .
Neuron-specific Wwox-knockout mice: These models allow researchers to study the specific effects of WWOX loss in neurons while maintaining expression in other tissues, helping to isolate neuronal contributions to the observed phenotypes .
Rat models: Rat models with Wwox deficiency have also been developed and show similar neurological phenotypes to mouse models, providing complementary systems for research .
The striking phenotypic similarity between rodent models and human patients (both showing severe epileptic seizures and growth retardation) makes these models particularly valuable for translational research .
Cellular models have been instrumental in elucidating WWOX's role in DNA repair:
MEF (Mouse Embryonic Fibroblast) lines: Comparison of Wwox-knockout and wild-type MEFs reveals that absence of Wwox is associated with mild genome instability, including chromosomal alterations and specific copy number variations (CNVs). Karyotype analysis of knockout MEFs showed near tetraploidy with structural abnormalities including del(7)(A1B4) and del(4)(C4) not present in wild-type cells .
Human cancer cell lines with modified WWOX expression:
These models enable detailed investigation of how WWOX affects different DNA repair pathways through techniques such as comet assays (for detecting DSBs), immunofluorescence assays for DSB markers (53BP1 and γH2AX), and pathway-specific reporter assays .
Brain MRI studies in patients with WWOX mutations reveal several consistent abnormalities:
Hypoplasia of the corpus callosum
Progressive cerebral atrophy
Delayed myelination
These neuroimaging findings correlate with the severe neurological phenotypes observed in affected individuals, particularly those with WOREE syndrome. The specific pattern of myelination defects seen in human patients is mirrored in mouse models, where hypomyelination is a characteristic feature . This consistency between human and mouse phenotypes supports the validity of mouse models for studying the molecular mechanisms underlying these developmental abnormalities.
Current treatment approaches for WWOX-related disorders show limited efficacy:
Antiepileptic drugs (AEDs): WOREE syndrome is generally refractory to current AEDs, representing a significant unmet medical need. In contrast, epilepsy in SCAR12 patients can be partially managed with available AEDs, though the ataxia and intellectual disability persist .
Emerging gene therapy approaches: Recent preclinical research has demonstrated that AAV9-mediated delivery of WWOX gene therapy (AAV-SynI-WWOX) can rescue multiple phenotypes in Wwox-null mice:
This proof-of-concept research suggests that neuronal restoration of WWOX expression through gene therapy could potentially treat both WOREE syndrome and SCAR12. The research was conducted using adeno-associated viral vectors (AAV9) carrying either murine Wwox or human WWOX cDNA under the control of the human neuronal Synapsin I promoter, ensuring neuron-specific expression .
WWOX plays a complex role in regulating DNA double-strand break (DSB) repair pathway choice, with significant implications for genome stability and cancer therapy resistance:
This pathway regulation suggests that WWOX status might predict tumor response to radiation therapy and certain chemotherapeutic agents, with potential applications in personalized cancer treatment strategies.
WWOX contributes to genome maintenance through several mechanisms:
Chromosomal stability: Karyotype analysis of Wwox-knockout MEFs revealed specific structural abnormalities not present in wild-type cells, including del(7)(A1B4) and del(4)(C4) .
Copy number variations (CNVs): Array comparative genomic hybridization analysis identified three distinct deletions shared by different Wwox-knockout MEF lines at chromosome locations 1 H6, 4 B3 and 8 C2, suggesting that Wwox absence leads to specific patterns of genomic instability .
Response to DNA damage: While Wwox deficiency doesn't appear to cause spontaneous DNA damage (as measured by comet assays and immunofluorescence for DSB markers), it does alter the cellular response to induced damage through its effects on repair pathway choice .
Tumor suppressor function: The location of WWOX in the chromosomal common fragile site FRA16D suggests its involvement in preventing genomic instability in regions prone to breakage under replication stress .
Understanding these mechanisms is crucial for interpreting how WWOX mutations contribute to both neurodevelopmental disorders and cancer predisposition.
Development of WWOX gene therapy requires addressing several technical considerations:
Vector design and selection:
AAV9 serotype has shown efficacy in crossing the blood-brain barrier and targeting neurons
The human neuronal Synapsin I promoter (SynI) provides neuron-specific expression, preventing off-target effects in other tissues
Vector capacity must accommodate the WWOX cDNA (~1.1 kb) along with regulatory elements
Delivery optimization:
Timing of intervention appears critical - neonatal administration was effective in mouse models, suggesting early intervention may be necessary for human patients
Route of administration (intracranial vs. intravenous) affects distribution and efficacy in the central nervous system
Dosing studies are needed to determine the minimum effective dose and therapeutic window
Safety considerations:
The proof-of-concept success in mouse models provides encouragement for further development, but translation to human patients will require additional preclinical studies in larger animal models and careful safety assessment.
Optimizing genetic testing strategies for WWOX-related disorders requires consideration of several factors:
Clinical predictors for targeted testing:
The incorporation of epilepsy genetics into clinical practice suggests that clinician prediction of specific genetic causes has limited sensitivity, arguing for parallel gene testing approaches rather than sequential single-gene testing .
For patients with early-onset epileptic encephalopathy, severe developmental delay, and brain abnormalities consistent with WOREE syndrome, WWOX should be included in first-line testing panels.
Testing methodologies:
Next-generation sequencing (NGS) panels for epilepsy genes, including WWOX
Whole exome sequencing (WES) for cases where targeted panels are negative
Copy number variation (CNV) analysis, as some WWOX-related disorders result from partial or complete gene deletions
RNA sequencing may be valuable for detecting splicing variants
Age-specific considerations:
Genetic counseling integration:
A multidisciplinary approach involving neurologists, geneticists, genetic counselors, and epileptologists provides the most comprehensive care pathway for patients with suspected WWOX-related disorders.
The WWOX gene was first identified through shotgun genomic sequencing and analysis of transcripts mapping to a region of chromosome 16 commonly affected by allelic loss in breast cancer . Researchers Bednarek et al. (2000) cloned the gene and named it WWOX due to its structure, which includes two WW domains and a region with high homology to the SRD family of enzymes . The gene was independently identified by Ried et al. (2000) and named FOR (fragile site FRA16D oxidoreductase) .
WWOX is expressed predominantly in hormonally regulated tissues such as the testis, ovary, and prostate . The protein plays a crucial role in various cellular functions, including protein degradation, transcription, and RNA splicing . The presence of WW domains suggests a role in protein-protein interactions, while the SRD domain indicates involvement in steroid metabolism .
WWOX has been shown to be an essential mediator of tumor necrosis factor-alpha-induced apoptosis . It binds directly to the p53 protein, a well-known tumor suppressor, and is involved in p53-mediated apoptosis . The high conservation of WWOX protein between humans and mice (93% identity) supports its significant role in apoptosis . Additionally, WWOX behaves as a potent suppressor of tumor growth, and abnormalities in this gene may contribute to carcinogenesis .
Mutations or inactivation of the WWOX gene have been associated with various human cancers, including breast cancer . Wwox null mice, which lack the WWOX gene, die prematurely, precluding adult tumor analysis . However, aging Wwox-heterozygous mice have been observed to develop a higher incidence of mammary tumors .