Khaled S. Abd-Elrahman

@ucalgary.ca

Smooth Muscle Research Group, Cumming School of Medicine
University of Calgary in Alberta



              

https://researchid.co/kshabdel
51

Scopus Publications

906

Scholar Citations

18

Scholar h-index

30

Scholar i10-index

Scopus Publications

  • Sexual dimorphism of G protein-coupled receptor signaling in the brain
    Sara Aljoudi, Hamdan Hamdan, and Khaled S. Abd-Elrahman

    Medknow

  • A M1 muscarinic acetylcholine receptor-specific positive allosteric modulator VU0486846 reduces neurogliosis in female Alzheimer's mice
    Khaled S. Abd-Elrahman, Tash-Lynn L. Colson, Shaarika Sarasija, and Stephen S.G. Ferguson

    Elsevier BV

  • Editorial: Emerging insights in glutamate receptor signaling in psychiatric, neurodevelopmental, and neurodegenerative diseases
    Khaled S. Abd-Elrahman, Jean-Martin Beaulieu, and Stephen S. G. Ferguson

    Frontiers Media SA

  • Targeting mGluR group III for the treatment of neurodegenerative diseases
    Nadia Rabeh, Baraa Hajjar, Jude O. Maraka, Ashwin F. Sammanasunathan, Mohammed Khan, Saif M.I. Alkhaaldi, Samy Mansour, Rashed T. Almheiri, Hamdan Hamdan, and Khaled S. Abd-Elrahman

    Elsevier BV

  • Comparison of Huntington’s disease phenotype progression in male and female heterozygous FDNQ175 mice
    Si Han Li, Tash-Lynn L. Colson, Jingwei Chen, Khaled S. Abd-Elrahman, and Stephen S. G. Ferguson

    Springer Science and Business Media LLC
    AbstractHuntington’s Disease (HD) is an inherited autosomal dominant neurodegenerative disorder that leads to progressive motor and cognitive impairment due to the expansion of a polyglutamine (CAG) repeat in the N-terminal region of the huntingtin (Htt) protein. The creation of HD mouse models represents a critical step in the research for HD treatment. Among the currently available HD mouse models, the zQ175 knock-in mouse line is the first to display robust disease phenotype on a heterozygous background. The newer FDNQ175 mouse model is derived from the zQ175 mouse line and presents a more aggressive phenotype. Moreover, increasing evidence has implicated sex as a contributing factor in the progression of HD symptoms. Here, we compared the progression of HD phenotypes in male and female heterozygous FDNQ175 mice. We found that both male and female heterozygous mice showed deficits in forelimb grip strength and cognition as early as 6 months of age. However, female FDNQ175 mice were less vulnerable to HD-associated decline in limb coordination and movement. Neither male nor female FDNQ175 mice exhibited reduced locomotor activity in the open field or exhibit consistent differences in anxiety at 6–12 months of age. Both male and female FDNQ175 mice exhibited increased numbers of huntingtin aggregates with age and 8-month-old female FDNQ175 mice had significantly more aggregates than their male counterparts. Taken together, our results provide further evidence that sex can influence the progression of HD phenotype in preclinical animal models and must be taken into consideration for future HD research.

  • VGLUT3 Deletion Rescues Motor Deficits and Neuronal Loss in the zQ175 Mouse Model of Huntington's Disease
    Karim S. Ibrahim, Salah El Mestikawy, Khaled S. Abd-Elrahman, and Stephen S.G. Ferguson

    Society for Neuroscience
    Huntington's disease (HD) is an autosomal-dominant neurodegenerative disease characterized by progressive motor and cognitive impairments, with no disease-modifying therapies yet available. HD pathophysiology involves evident impairment in glutamatergic neurotransmission leading to severe striatal neurodegeneration. The vesicular glutamate transporter-3 (VGLUT3) regulates the striatal network that is centrally affected by HD. Nevertheless, current evidence on the role of VGLUT3 in HD pathophysiology is lacking. Here, we crossed mice lackingSlc17a8gene (VGLUT3–/–) with heterozygouszQ175knock-in mouse model of HD (zQ175:VGLUT3–/–). Longitudinal assessment of motor and cognitive functions from 6 to 15 months of age reveals that VGLUT3 deletion rescues motor coordination and short-term memory deficits in both male and femalezQ175mice. VGLUT3 deletion also rescues neuronal loss likely via the activation of Akt and ERK1/2 in the striatum ofzQ175mice of both sexes. Interestingly, the rescue in neuronal survival inzQ175:VGLUT3–/–mice is accompanied by a reduction in the number of nuclear mutant huntingtin (mHTT) aggregates with no change in the total aggregate levels or microgliosis. Collectively, these findings provide novel evidence that VGLUT3, despite its limited expression, can be a vital contributor to HD pathophysiology and a viable target for HD therapeutics.SIGNIFICANCE STATEMENTDysregulation of the striatal network centrally contributes to the pathophysiology of Huntington's disease (HD). The atypical vesicular glutamate transporter-3 (VGLUT3) has been shown to regulate several major striatal pathologies, such as addiction, eating disorders, or L-DOPA-induced dyskinesia. Yet, our understanding of VGLUT3's role in HD remains unclear. We report here that deletion of theSlc17a8(Vglut3) gene rescues the deficits in both motor and cognitive functions in HD mice of both sexes. We also find that VGLUT3 deletion activates neuronal survival signaling and reduces nuclear aggregation of abnormal huntingtin proteins and striatal neuron loss in HD mice. Our novel findings highlight the vital contribution of VGLUT3 in HD pathophysiology that can be exploited for HD therapeutic management.

  • The Role of Neuroglial Metabotropic Glutamate Receptors in Alzheimer’s Disease
    Khaled S. Abd-Elrahman, Stephen S.G. Ferguson, and Shaarika Sarasija

    Bentham Science Publishers Ltd.
    : Glutamate, the major excitatory neurotransmitter in the brain exerts its effects via both ionotropic glutamate receptors and metabotropic glutamate receptors (mGluRs). There are three subgroups of mGluRs, pre-synaptic Group II and Group III mGluRs and post-synaptic Group I mGluRs. mGluRs are ubiquitously expressed in the brain and their activation is poised upstream of a myriad of signaling pathways, resulting in their implication in the pathogenesis of various neurodegenerative diseases including, Alzheimer’s Disease (AD). While the exact mechanism of AD etiology remains elusive, β-amyloid (Aβ) plaques and hyperphosphorylated tau tangles remain the histopathological hallmarks of AD. Though less electrically excitable, neuroglia are a major non-neuronal cell type in the brain and are composed of astrocytes, microglia, and oligodendrocytes. Astrocytes, microglia, and oligodendrocytes provide structural and metabolic support, active immune defence, and axonal support and sheathing, respectively. Interestingly, Aβ and hyperphosphorylated tau are known to disrupt the neuroglial homeostasis in the brain, pushing them towards a more neurotoxic state. In this review, we discuss what is currently known regarding the expression patterns of various mGluRs in neuroglia and how Aβ and tau alter the normal mGluR function in the neuroglia and contribute to the pathophysiology of AD.

  • Adipose tissue mitochondrial dysfunction and cardiometabolic diseases: On the search for novel molecular targets
    Ibrahim AlZaim, Ali H. Eid, Khaled S. Abd-Elrahman, and Ahmed F. El-Yazbi

    Elsevier BV

  • Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases
    Si Han Li, Khaled S. Abd-Elrahman, and Stephen S.G. Ferguson

    Elsevier BV

  • Editorial: Methods and application in cardiovascular and smooth muscle pharmacology: 2021
    Ahmed F. El-Yazbi, Ali H. Eid, Fouad A. Zouein, and Khaled S. Abd-Elrahman

    Frontiers Media SA
    Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt, Faculty of Pharmacy, Alamein International University, Al Alamein, Egypt, Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar, Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon, Department of Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Inserm, Université ParisSaclay, Paris-Saclay, France, The Cardiovascular Renal and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Beirut, Lebanon, Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States, Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates

  • VGLUT3 Ablation Differentially Modulates Glutamate Receptor Densities in Mouse Brain
    Karim S. Ibrahim, Salah El Mestikawy, Khaled S. Abd-Elrahman, and Stephen S.G. Ferguson

    Society for Neuroscience
    Abstract Type 3 vesicular glutamate transporter (VGLUT3) represents a unique modulator of glutamate release from both nonglutamatergic and glutamatergic varicosities within the brain. Despite its limited abundance, VGLUT3 is vital for the regulation of glutamate signaling and, therefore, modulates the activity of various brain microcircuits. However, little is known about how glutamate receptors are regulated by VGLUT3 across different brain regions. Here, we used VGLUT3 constitutive knock-out (VGLUT3–/–) mice and explored how VGLUT3 deletion influences total and cell surface expression of different ionotropic and metabotropic glutamate receptors. VGLUT3 deletion upregulated the overall expression of metabotropic glutamate receptors mGluR5 and mGluR2/3 in the cerebral cortex. In contrast, no change in the total expression of ionotropic NMDAR glutamate receptors were observed in the cerebral cortex of VGLUT3–/– mice. We noted significant reduction in cell surface levels of mGluR5, NMDAR2A, NMDAR2B, as well as reductions in dopaminergic D1 receptors and muscarinic M1 acetylcholine receptors in the hippocampus of VGLUT3–/– mice. Furthermore, mGluR2/3 total expression and mGluR5 cell surface levels were elevated in the striatum of VGLUT3–/– mice. Last, AMPAR subunit GluA1 was significantly upregulated throughout cortical, hippocampal, and striatal brain regions of VGLUT3–/– mice. Together, these findings complement and further support the evidence that VGLUT3 dynamically regulates glutamate receptor densities in several brain regions. These results suggest that VGLUT3 may play an intricate role in shaping glutamatergic signaling and plasticity in several brain areas.

  • Early metabolic impairment as a contributor to neurodegenerative disease: Mechanisms and potential pharmacological intervention
    Walaa Fakih, Ralph Zeitoun, Ibrahim AlZaim, Ali H. Eid, Firas Kobeissy, Khaled S. Abd‐Elrahman, and Ahmed F. El‐Yazbi

    Wiley
    The metabolic syndrome comprises a family of clinical and laboratory findings, including insulin resistance, hyperglycemia, hypertriglyceridemia, low high‐density lipoprotein cholesterol levels, and hypertension, in addition to central obesity. The syndrome confers a high risk of cardiovascular mortality. Indeed, metabolic dysfunction has been shown to cause a direct insult to smooth muscle and endothelial components of the vasculature, which leads to vascular dysfunction and hyperreactivity. This, in turn, causes cerebral vasoconstriction and hypoperfusion, eventually contributing to cognitive deficits. Moreover, the metabolic syndrome disrupts key homeostatic processes in the brain, including apoptosis, autophagy, and neurogenesis. Impairment of such processes in the context of metabolic dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, including Alzheimer, Parkinson, and Huntington diseases.

  • A positive allosteric modulator for the muscarinic receptor (M1 mAChR) improves pathology and cognitive deficits in female APPswe/PSEN1ΔE9 mice
    Khaled S. Abd‐Elrahman, Shaarika Sarasija, Tash‐Lynn L. Colson, and Stephen S. G. Ferguson

    Wiley
    BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline and women account for 60% of diagnosed cases. Beta-amyloid (Aβ) oligomers is considered the principal neurotoxic species in AD brains. The M1 muscarinic acetylcholine receptor (M1 mAChR) plays a key role in memory and learning. M1 mAChR agonists show pro-cognitive activity but cause many adverse off-target effects. A new orally bioavailable M1 mAChR positive allosteric modulator (PAM), VU0486846, is devoid of direct agonist activity or adverse effects but was not tested for disease-modifying efficacy in female AD mice. EXPERIMENTAL APPROACH Nine-month-old female APPswe/PSEN1ΔE9 (APPswe) and wildtype mice were treated with VU0486846 in drinking water (10mg/kg/day) for 4 or 8 weeks. Cognitive function of mice was assessed after treatment and brains were harvested for biochemical and immunohistochemical assessment. KEY RESULTS VU0486846 improved cognitive function of APPswe mice when tested in novel object recognition and Morris water maze. This was paralleled by a significant reduction in Aβ oligomers and plaques and neuronal loss in hippocampus. VU0486846 reduced Aβ oligomer production in APPswe mice by increasing M1 mAChR expression and shifting the processing of amyloid precursor protein from amyloidogenic cleavage to non-amyloidogenic cleavage. Specifically, VU0486846 reduced the expression of β-secretase 1 (BACE1), whereas it enhanced the expression of the α-secretase ADAM10 in APPswe hippocampus. CONCLUSION AND IMPLICATIONS Using M1 mAChR PAMs can be a viable disease-modifying approach that should be exploited clinically to slow AD in women.

  • Metabotropic Glutamate Receptor 5 Antagonism Reduces Pathology and Differentially Improves Symptoms in Male and Female Heterozygous zQ175 Huntington’s Mice
    Si Han Li, Tash-Lynn L. Colson, Khaled S. Abd-Elrahman, and Stephen S. G. Ferguson

    Frontiers Media SA
    Huntington’s disease (HD) is an inherited autosomal dominant neurodegenerative disorder that leads to progressive motor and cognitive impairment. There are currently no available disease modifying treatments for HD patients. We have previously shown that pharmacological blockade of metabotropic glutamate receptor 5 (mGluR5) signaling rescues motor deficits, improves cognitive impairments and mitigates HD neuropathology in male zQ175 HD mice. Mounting evidence indicates that sex may influence HD progression and we have recently reported a sex-specific pathological mGluR5 signaling in Alzheimer’s disease (AD) mice. Here, we compared the outcomes of treatment with the mGluR5 negative allosteric modulator CTEP (2-chloro-4-[2-[2,5-dimethyl-1-[4-(trifluoromethoxy)phenyl]imidazol-4-yl]ethynyl]pyridine) in both male and female symptomatic zQ175 mice. We found that female zQ175 mice required a longer treatment duration with CTEP than male mice to show improvement in their rotarod performance. Unlike males, chronic CTEP treatment did not improve the grip strength nor reverse the cognitive decline of female zQ175 mice. However, CTEP reduced the number of huntingtin aggregates, improved neuronal survival and decreased microglia activation in the striatum of both male and female zQ175 mice. Together, our results indicate that mGluR5 antagonism can reduce HD neuropathology in both male and female zQ175 HD mice, but sex has a clear impact on the efficacy of the treatment and must be taken into consideration for future HD drug development.

  • Noncanonical Metabotropic Glutamate Receptor 5 Signaling in Alzheimer's Disease
    Khaled S. Abd-Elrahman and Stephen S.G. Ferguson

    Annual Reviews
    Metabotropic glutamate receptor 5 (mGluR5) is ubiquitously expressed in brain regions responsible for memory and learning. It plays a key role in modulating rapid changes in synaptic transmission and plasticity. mGluR5 supports long-term changes in synaptic strength by regulating the transcription and translation of essential synaptic proteins. β-Amyloid 42 (Aβ42) oligomers interact with a mGluR5/cellular prion protein (PrPC) complex to disrupt physiological mGluR5 signal transduction. Aberrant mGluR5 signaling and associated synaptic failure are considered an emerging pathophysiological mechanism of Alzheimer's disease (AD). Therefore, mGluR5 represents an attractive therapeutic target for AD, and recent studies continue to validate the efficacy of various mGluR5 allosteric modulators in improving memory deficits and mitigating disease pathology. However, sex-specific differences in the pharmacology of mGluR5 and activation of noncanonical signaling downstream of the receptor suggest that its utility as a therapeutic target in female AD patients needs to be reconsidered.

  • Metabotropic Glutamate Receptor 2/3 Activation Improves Motor Performance and Reduces Pathology in Heterozygous zQ175 Huntington Disease Mice
    Si Han Li, Tash-Lynn L. Colson, Khaled S. Abd-Elrahman, and Stephen S.G. Ferguson

    American Society for Pharmacology & Experimental Therapeutics (ASPET)
    Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease that leads to progressive motor impairments with no available disease-modifying treatment. Current evidence indicates that exacerbated postsynaptic glutamate signaling in the striatum plays a key role in the pathophysiology of HD. However, it remains unclear whether reducing glutamate release can be an effective approach to slow the progression of HD. Here, we show that the activation of metabotropic glutamate receptors 2 and 3 (mGluR2/3), which inhibit presynaptic glutamate release, improves HD symptoms and pathology in heterozygous zQ175 knockin mice. Treatment of both male and female zQ175 mice with the potent and selective mGluR2/3 agonist LY379268 for either 4 or 8 weeks improves both limb coordination and locomotor function in all mice. LY379268 also reduces mutant huntingtin aggregate formation, neuronal cell death, and microglial activation in the striatum of both male and female zQ175 mice. The reduction in mutant huntingtin aggregates correlates with the activation of a glycogen synthase kinase 3β–dependent autophagy pathway in male, but not female, zQ175 mice. Furthermore, LY379268 reduces both Akt and ERK1/2 phosphorylation in male zQ175 mice but increases both Akt and ERK1/2 phosphorylation in female zQ175 mice. Taken together, our results indicate that mGluR2/3 activation mitigates HD neuropathology in both male and female mice but is associated with the differential activation and inactivation of cell signaling pathways in heterozygous male and female zQ175 mice. This further highlights the need to take sex into consideration when developing future HD therapeutics. SIGNIFICANCE STATEMENT The mGluR2/3 agonist LY379268 improves motor impairments and reduces pathology in male and female zQ175 Huntington’s disease mice. The beneficial outcomes of LY379268 treatment in Huntington’s disease mice were mediated by divergent cell signaling pathways in both sexes. We provide evidence that mGluR2/3 agonists can be repurposed for the treatment of Huntington’s disease, and we emphasize the importance of investigating sex as a biological variable in preclinical disease-modifying studies.

  • Optineurin deletion disrupts metabotropic glutamate receptor 5-mediated regulation of ERK1/2, GSK3β/ZBTB16, mTOR/ULK1 signaling in autophagy
    Karim S. Ibrahim, Caitlyn J. McLaren, Khaled S. Abd-Elrahman, and Stephen S.G. Ferguson

    Elsevier BV
    Optineurin (OPTN) is a multifunctional protein that mediates a network of cellular processes regulating membrane trafficking, inflammatory responses and autophagy. The OPTN-rich interactome includes Group I metabotropic glutamate receptors (mGluR1 and 5), members of the Gαq/11 protein receptor family. Recent evidence has shown that mGluR5, in addition to its canonical Gαq/11 protein-coupled signaling, regulates autophagic machinery via mTOR/ULK1 and GSK3β/ZBTB16 pathways in both Alzheimer's and Huntington's disease mouse models. Despite its potential involvement, the role of OPTN in mediating mGluR5 downstream signaling cascades remains largely unknown. Here, we employed a CRISPR/Cas9 OPTN-deficient STHdhQ7/Q7 striatal cell line and global OPTN knockout mice to investigate whether Optn gene deletion alters both mGluR5 canonical and noncanonical signaling. We find that OPTN is required for mGluR5-activated Ca2+ flux and ERK1/2 signaling following receptor activation in STHdhQ7/Q7 cells and acute hippocampal slices. Deletion of OPTN impairs both GSK3β/ZBTB16 and mTOR/ULK1 autophagic signaling in STHdhQ7/Q7 cells. Furthermore, mGluR5-dependent regulation of GSK3β/ZBTB16 and mTOR/ULK-1 autophagic signaling is impaired in hippocampal slices of OPTN knockout mice. Overall, we show that the crosstalk between OPTN and mGluR5 can have major implication on receptor signaling and therefore potentially contribute to the pathophysiology of neurodegenerative diseases.

  • Experimental investigation of rc columns confined with ni–ti shape memory alloy wires versus cfrp sheets
    Khaled Abdelrahman and Raafat El-Hacha

    Canadian Science Publishing
    The critical need to enhance existing strengthening methods with more efficient and effective ones has led to the evolvement of smarter and innovative class of materials termed shape memory alloys (SMA). The SMAs possess unique characteristic properties that lie in their ability to undergo large deformations and return to their undeformed shape through stress removal or heating process. Limited research studies conducted using SMAs have shown high potential for their use in building industry. Results presented in this research study are from an experimental study that investigated the compressive behaviour of uniaxial concentrically loaded nickel–titanium (Ni–Ti) SMA-spirally confined RC columns and compared with RC columns confined with conventional carbon fibre reinforced polymer (CFRP) sheets. The compression tests revealed that actively confining the concrete column with Ni–Ti SMA spiral wires increased the performance of the concrete dramatically. Additionally, the active Ni–Ti SMA-confinement system exhibited superior performance compared to the conventional passive CFRP-confinement system.

  • The pleiotropic effects of antithrombotic drugs in the metabolic-cardiovascular-neurodegenerative disease continuum: Impact beyond reduced clotting
    Rana A. Alaaeddine, Ibrahim AlZaim, Safaa H. Hammoud, Aya Arakji, Ali H. Eid, Khaled S. Abd-Elrahman, and Ahmed F. El-Yazbi

    Portland Press Ltd.
    Abstract Antithrombotic drugs are widely used for primary and secondary prevention, as well as treatment of many cardiovascular disorders. Over the past few decades, major advances in the pharmacology of these agents have been made with the introduction of new drug classes as novel therapeutic options. Accumulating evidence indicates that the beneficial outcomes of some of these antithrombotic agents are not solely related to their ability to reduce thrombosis. Here, we review the evidence supporting established and potential pleiotropic effects of four novel classes of antithrombotic drugs, adenosine diphosphate (ADP) P2Y12-receptor antagonists, Glycoprotein IIb/IIIa receptor Inhibitors, and Direct Oral Anticoagulants (DOACs), which include Direct Factor Xa (FXa) and Direct Thrombin Inhibitors. Specifically, we discuss the molecular evidence supporting such pleiotropic effects in the context of cardiovascular disease (CVD) including endothelial dysfunction (ED), atherosclerosis, cardiac injury, stroke, and arrhythmia. Importantly, we highlight the role of DOACs in mitigating metabolic dysfunction-associated cardiovascular derangements. We also postulate that DOACs modulate perivascular adipose tissue inflammation and thus, may reverse cardiovascular dysfunction early in the course of the metabolic syndrome. In this regard, we argue that some antithrombotic agents can reverse the neurovascular damage in Alzheimer’s and Parkinson’s brain and following traumatic brain injury (TBI). Overall, we attempt to provide an up-to-date comprehensive review of the less-recognized, beneficial molecular aspects of antithrombotic therapy beyond reduced thrombus formation. We also make a solid argument for the need of further mechanistic analysis of the pleiotropic effects of antithrombotic drugs in the future.

  • Aβ oligomers induce pathophysiological mGluR5 signaling in Alzheimer's disease model mice in a sex-selective manner
    Khaled S. Abd-Elrahman, Awatif Albaker, Jessica M. de Souza, Fabiola M. Ribeiro, Michael G. Schlossmacher, Mario Tiberi, Alison Hamilton, and Stephen S. G. Ferguson

    American Association for the Advancement of Science (AAAS)
    A sex-specific prion-amyloid interaction predicts that some Alzheimer’s disease therapies may not work in female patients. Men only for an Alzheimer’s drug target β-Amyloid (Aβ) deposits in the brain contribute to the progression of Alzheimer’s disease (AD) by inducing excitotoxic signaling in neurons. Aβ binds to the metabotropic glutamate receptor mGluR5, and pharmacological inhibition of mGluR5 reverses cognitive decline in male AD model animals. However, Abd-Elrahman et al. found that Aβ bound to mGluR5 in postmortem brain tissue only from male AD model mice and male human donors. This sex-selective interaction was mediated by prion protein in male mouse brain tissue. The findings suggest that mGluR5 inhibitors may be therapeutically beneficial only for male patients with AD. The prevalence, presentation, and progression of Alzheimer’s disease (AD) differ between men and women, although β-amyloid (Aβ) deposition is a pathological hallmark of AD in both sexes. Aβ-induced activation of the neuronal glutamate receptor mGluR5 is linked to AD progression. However, we found that mGluR5 exhibits distinct sex-dependent profiles. Specifically, mGluR5 isolated from male mouse cortical and hippocampal tissues bound with high affinity to Aβ oligomers, whereas mGluR5 from female mice exhibited no such affinity. This sex-selective Aβ-mGluR5 interaction did not appear to depend on estrogen, but rather Aβ interaction with cellular prion protein (PrPC), which was detected only in male mouse brain homogenates. The ternary complex between mGluR5, Aβ oligomers, and PrPC was essential to elicit mGluR5-dependent pathological suppression of autophagy in primary neuronal cultures. Pharmacological inhibition of mGluR5 reactivated autophagy, mitigated Aβ pathology, and reversed cognitive decline in male APPswe/PS1ΔE9 mice, but not in their female counterparts. Aβ oligomers also bound with high affinity to human mGluR5 isolated from postmortem donor male cortical brain tissue, but not that from female samples, suggesting that this mechanism may be relevant to patients. Our findings indicate that mGluR5 does not contribute to Aβ pathology in females, highlighting the complexity of mGluR5 pharmacology and Aβ signaling that supports the need for sex-specific stratification in clinical trials assessing AD therapeutics.

  • Targeting vesicular glutamate transporter machinery: Implications on metabotropic glutamate receptor 5 signaling and behavior
    Karim S. Ibrahim, Khaled S. Abd-Elrahman, Salah El Mestikawy, and Stephen SG Ferguson

    American Society for Pharmacology & Experimental Therapeutics (ASPET)
    Crosstalk between both pre- and post-synaptic components of glutamatergic neurotransmission plays a crucial role in orchestrating a multitude of brain functions including synaptic plasticity and motor planning. Metabotropic glutamate receptor 5 (mGluR5) exhibits a promising therapeutic potential for many neurodevelopmental and neurodegenerative disorders, as the consequence of its modulatory control over diverse neuronal networks required for memory, motor coordination, neuronal survival and differentiation. Given these crucial roles, mGluR5 signaling is under the tight control of glutamate release machinery mediated through vesicular glutamate transporters (VGLUTs) to ultimately dictate glutamatergic output. A particular VGLUT isoform, VGLUT3, exhibits an overlapping, but unique, distribution with mGluR5 and the dynamic crosstalk between mGluR5 and VGLUT3 is key for the function of specific neuronal networks involved in motor coordination, emotions and cognition. Thus, aberrant signaling of the VGLUT3/mGluR5 axis is linked to various pathologies including, but not limited to, Parkinson's disease, anxiety disorders and drug addiction. We argue that a comprehensive profiling of how coordinated VGLUT3/mGluR5 signaling influences overall glutamatergic neurotransmission is warranted. Significance Statement Vesicular glutamate receptor 3 (VGLUT3) machinery orchestrates glutamate release and its distribution overlaps with metabotropic glutamate receptor 5 (mGluR5) in regional brain circuitries including striatum, hippocampus and raphe nucleus. Therefore, VGLUT3/mGluR5 crosstalk can significantly influences both physiological and pathophysiological glutamatergic neurotransmission. Pathological signaling of the VGLUT3/mGluR5 axis is linked to Parkinson's disease, anxiety disorders and drug addiction. However, it is also predicted to contribute to other motor and cognitive disorders.

  • MGluR5 regulates REST/NRSF signaling through N-cadherin/β-catenin complex in Huntington's disease
    Jéssica M. de Souza, Khaled S. Abd-Elrahman, Fabiola M. Ribeiro, and Stephen S. G. Ferguson

    Springer Science and Business Media LLC
    AbstractRepressor element 1-silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is a transcription repressor and its expression is regulated by the Wnt pathway through β-catenin. Metabotropic glutamate receptor 5 (mGluR5) signaling plays a key role in controlling neuronal gene expression. Interestingly, REST/NRSF nuclear translocation and signaling, as well as mGluR5 signaling are altered in the presence of mutant huntingtin. It remains unclear whether mGluR5 can modulate Wnt and REST/NRSF signaling under physiological conditions and whether this modulation is altered in Huntington’s disease (HD). Using primary corticostriatal neurons derived from wild type mouse embryos, we find that targeting mGluR5 using the agonist, DHPG, or the negative allosteric modulator, CTEP, modulates REST/NRSF expression by regulating the assembly of N-cadherin/ β-catenin complex in a Src kinase-dependent manner. We have validated our in vitro findings in vivo using two HD mouse models. Specifically, we show that pharmacological inhibition of mGluR5 in zQ175 mice and genetic ablation of mGluR5 in BACHD mice corrected the pathological activation of Src and rescued REST/NRSF-dependent signaling. Together, our data provide evidence that mGluR5 regulates REST/NRSF expression via the Wnt pathway and highlight the contribution of impaired REST/ NRSF signaling to HD pathology.



  • MGluR5 Contribution to Neuropathology in Alzheimer Mice Is Disease Stage-Dependent
    Khaled S. Abd-Elrahman, Alison Hamilton, Awatif Albaker, and Stephen S. G. Ferguson

    American Chemical Society (ACS)
    Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is characterized by a progressive cognitive decline in affected individuals. Current therapeutic strategies are limited in their efficacy and some have proven to be even less effective at later disease stages or after extended use. We previously demonstrated that chronic inhibition of mGluR5 signaling using the selective negative allosteric modulator (NAM) CTEP in APPswe/PS1ΔE9 mice can rescue cognitive function, activating the ZBTB16-mediated autophagy pathway to reduce Aβ, the principal neurotoxic species in AD brains. Here, we evaluated the efficacy of long-term treatment with CTEP in 6 month old APPswe/PS1ΔE9 mice for either 24 or 36 weeks. CTEP maintained its efficacy in reversing working and spatial memory deficits and mitigating neurogliosis in APPswe/PS1ΔE9 mice when administered for 24 weeks. This was paralleled by a significant reduction in Aβ oligomer and plaque load as a result of autophagy activation via ZBTB16 and mTOR-dependent pathways. However, further extension of CTEP treatment for 36 weeks was found ineffective in reversing memory deficit, neurogliosis, or Aβ-related pathology. We found that this loss in CTEP efficacy in 15 month old APPswe/PS1ΔE9 mice was due to the abolished contribution of ZBTB16 and mTOR-mediated signaling to AD neuropathology at this advanced disease stage. Our findings indicate that the contribution of pathological mGluR5-signaling to AD may shift as the disease progresses. Thus, we provide the first evidence that the underlying pathophysiological mechanism(s) of AD may unfold along the course of the disease and treatment strategies should be modified accordingly to ensure maximal therapeutic outcomes.

RECENT SCHOLAR PUBLICATIONS

  • 大脑中 G 蛋白偶联受体信号传导的两性异形
    S Aljoudi, H Hamdan, KS Abd-Elrahman
    中国神经再生研究 (英文版) 19 (8), 1635 2024

  • Sexual dimorphism of G protein-coupled receptor signaling in the brain
    S Aljoudi, H Hamdan, KS Abd-Elrahman
    Neural Regeneration Research 19 (8), 1635-1636 2024

  • A M1 muscarinic acetylcholine receptor-specific positive allosteric modulator VU0486846 reduces neurogliosis in female Alzheimer’s mice
    KS Abd-Elrahman, TLL Colson, S Sarasija, SSG Ferguson
    Biomedicine & Pharmacotherapy 173, 116388 2024

  • Emerging insights in glutamate receptor signaling in psychiatric, neurodevelopmental, and neurodegenerative diseases
    KS Abd-Elrahman, JM Beaulieu, SSG Ferguson
    Frontiers in Cell and Developmental Biology 12, 1388514 2024

  • Targeting mGluR group III for the treatment of neurodegenerative diseases
    N Rabeh, B Hajjar, JO Maraka, AF Sammanasunathan, M Khan, ...
    Biomedicine & Pharmacotherapy 168, 115733 2023

  • Comparison of Huntington’s disease phenotype progression in male and female heterozygous FDNQ175 mice
    SH Li, TLL Colson, J Chen, KS Abd-Elrahman, SSG Ferguson
    Molecular Brain 16 (1), 67 2023

  • VGLUT3 deletion rescues motor deficits and neuronal loss in the zQ175 mouse model of Huntington's disease
    KS Ibrahim, S El Mestikawy, KS Abd-Elrahman, SSG Ferguson
    Journal of Neuroscience 43 (23), 4365-4377 2023

  • Genetic deletion of atypical VGLUT3 rescues Huntington’s disease phenotype and neurodegeneration in zQ175 mice
    K Ibrahim, K Abd-Elrahman, SS Ferguson
    Journal of Pharmacology and Experimental Therapeutics 385 (S3) 2023

  • A positive allosteric modulator of M1 Acetylcholine receptors improves cognitive deficits in male and female APPswe/PSEN1ΔE9 mice via divergent mechanisms
    K Abd-Elrahman, TLL Colson, SS Ferguson
    Journal of Pharmacology and Experimental Therapeutics 385 (S3) 2023

  • The role of neuroglial metabotropic glutamate receptors in Alzheimer’s disease
    KS Abd-Elrahman, S Sarasija, SSG Ferguson
    Current Neuropharmacology 21 (2), 273 2023

  • Adipose tissue mitochondrial dysfunction and cardiometabolic diseases: On the search for novel molecular targets
    I AlZaim, AH Eid, KS Abd-Elrahman, AF El-Yazbi
    Biochemical Pharmacology 206, 115337 2022

  • Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases
    SH Li, KS Abd-Elrahman, SSG Ferguson
    Pharmacology & Therapeutics 239, 108275 2022

  • Methods and application in cardiovascular and smooth muscle pharmacology: 2021
    AF El-Yazbi, AH Eid, FA Zouein, KS Abd-Elrahman
    Frontiers in Pharmacology 13, 1049022 2022

  • Early metabolic impairment as a contributor to neurodegenerative disease: Mechanisms and potential pharmacological intervention
    W Fakih, R Zeitoun, I AlZaim, AH Eid, F Kobeissy, KS Abd‐Elrahman, ...
    Obesity 30 (5), 982-993 2022

  • VGLUT3 ablation differentially modulates glutamate receptor densities in mouse brain
    KS Ibrahim, S El Mestikawy, KS Abd-Elrahman, SSG Ferguson
    ENeuro 9 (3) 2022

  • A positive allosteric modulator for the muscarinic receptor (M1 mAChR) improves pathology and cognitive deficits in female APPswe/PSEN1ΔE9 mice
    KS Abd‐Elrahman, S Sarasija, TLL Colson, SSG Ferguson
    British Journal of Pharmacology 179 (8), 1769-1783 2022

  • Metabotropic glutamate receptor 5 antagonism reduces pathology and differentially improves symptoms in male and female heterozygous zQ175 Huntington’s mice
    SH Li, TLL Colson, KS Abd-Elrahman, SSG Ferguson
    Frontiers in Molecular Neuroscience 15, 801757 2022

  • Noncanonical metabotropic glutamate receptor 5 signaling in Alzheimer's disease
    KS Abd-Elrahman, SSG Ferguson
    Annual Review of Pharmacology and Toxicology 62, 235-254 2022

  • Metabotropic glutamate receptor 2/3 activation improves motor performance and reduces pathology in heterozygous zQ175 Huntington disease mice
    SH Li, TLL Colson, KS Abd-Elrahman, SSG Ferguson
    Journal of Pharmacology and Experimental Therapeutics 379 (1), 74-84 2021

  • Ablation of optineurin impairs metabotropic glutamate receptor 5 signaling in mouse hippocampus
    K Ibrahim, C McLaren, K Abd‐Elrahman, S Ferguson
    The FASEB Journal 35 2021

MOST CITED SCHOLAR PUBLICATIONS

  • mGluR5 antagonism increases autophagy and prevents disease progression in the zQ175 mouse model of Huntington’s disease
    KS Abd-Elrahman, A Hamilton, SR Hutchinson, F Liu, RC Russell, ...
    Science signaling 10 (510), eaan6387 2017
    Citations: 80

  • Stromatoxin‐sensitive, heteromultimeric Kv2. 1/Kv9. 3 channels contribute to myogenic control of cerebral arterial diameter
    XZ Zhong, KS Abd‐Elrahman, CH Liao, AF El‐Yazbi, EJ Walsh, MP Walsh, ...
    The Journal of physiology 588 (22), 4519-4537 2010
    Citations: 68

  • Aβ oligomers induce pathophysiological mGluR5 signaling in Alzheimer’s disease model mice in a sex-selective manner
    KS Abd-Elrahman, A Albaker, JM de Souza, FM Ribeiro, ...
    Science signaling 13 (662), eabd2494 2020
    Citations: 61

  • Modulation of mTOR and CREB pathways following mGluR5 blockade contribute to improved Huntington’s pathology in zQ175 mice
    KS Abd-Elrahman, SSG Ferguson
    Molecular brain 12 (1), 35 2019
    Citations: 56

  • Autophagy is increased following either pharmacological or genetic silencing of mGluR5 signaling in Alzheimer’s disease mouse models
    KS Abd-Elrahman, A Hamilton, M Vasefi, SSG Ferguson
    Molecular Brain 11, 1-8 2018
    Citations: 52

  • Pioglitazone abrogates cyclosporine-evoked hypertension via rectifying abnormalities in vascular endothelial function
    MM El-Mas, HM El-Gowelli, KS Abd-Elrahman, EI Saad, AGA Abdel-Galil, ...
    Biochemical pharmacology 81 (4), 526-533 2011
    Citations: 46

  • Noncanonical metabotropic glutamate receptor 5 signaling in Alzheimer's disease
    KS Abd-Elrahman, SSG Ferguson
    Annual Review of Pharmacology and Toxicology 62, 235-254 2022
    Citations: 40

  • mGluR5 contribution to neuropathology in Alzheimer mice is disease stage-dependent
    KS Abd-Elrahman, A Hamilton, A Albaker, SSG Ferguson
    ACS pharmacology & translational science 3 (2), 334-344 2020
    Citations: 40

  • PKC-mediated cerebral vasoconstriction: Role of myosin light chain phosphorylation versus actin cytoskeleton reorganization
    AF El-Yazbi, KS Abd-Elrahman, A Moreno-Dominguez
    Biochemical pharmacology 95 (4), 263-278 2015
    Citations: 39

  • Amelioration of perivascular adipose inflammation reverses vascular dysfunction in a model of nonobese prediabetic metabolic challenge: potential role of antidiabetic drugs
    MAW Elkhatib, A Mroueh, RW Rafeh, F Sleiman, H Fouad, EI Saad, ...
    Translational Research 214, 121-143 2019
    Citations: 33

  • Podocyte NADPH oxidase 5 promotes renal inflammation regulated by the toll-like receptor pathway
    CE Holterman, NC Boisvert, JF Thibodeau, E Kamto, M Novakovic, ...
    Antioxidants & redox signaling 30 (15), 1817-1830 2019
    Citations: 32

  • ROK and arteriolar myogenic tone generation: molecular evidence in health and disease
    AF El-Yazbi, KS Abd-Elrahman
    Frontiers in pharmacology 8, 245203 2017
    Citations: 32

  • mGluR5 allosteric modulation promotes neurorecovery in a 6-OHDA-toxicant model of Parkinson’s disease
    K Farmer, KS Abd-Elrahman, A Derksen, EM Rowe, AM Thompson, ...
    Molecular Neurobiology 57, 1418-1431 2020
    Citations: 31

  • Abnormal Rho-associated kinase activity contributes to the dysfunctional myogenic response of cerebral arteries in type 2 diabetes
    KS Abd-Elrahman, MP Walsh, WC Cole
    Canadian Journal of Physiology and Pharmacology 93 (3), 177-184 2015
    Citations: 25

  • Abnormal myosin phosphatase targeting subunit 1 phosphorylation and actin polymerization contribute to impaired myogenic regulation of cerebral arterial diameter in the type 2
    KS Abd-Elrahman, O Colinas, EJ Walsh, HL Zhu, CM Campbell, ...
    Journal of Cerebral Blood Flow & Metabolism 37 (1), 227-240 2017
    Citations: 22

  • mGluR5 regulates REST/NRSF signaling through N-cadherin/β-catenin complex in Huntington’s disease
    JM de Souza, KS Abd-Elrahman, FM Ribeiro, SSG Ferguson
    Molecular brain 13, 1-15 2020
    Citations: 21

  • A positive allosteric modulator for the muscarinic receptor (M1 mAChR) improves pathology and cognitive deficits in female APPswe/PSEN1ΔE9 mice
    KS Abd‐Elrahman, S Sarasija, TLL Colson, SSG Ferguson
    British Journal of Pharmacology 179 (8), 1769-1783 2022
    Citations: 20

  • Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases
    SH Li, KS Abd-Elrahman, SSG Ferguson
    Pharmacology & Therapeutics 239, 108275 2022
    Citations: 19

  • Targeting vesicular glutamate transporter machinery: implications on metabotropic glutamate receptor 5 signaling and behavior
    KS Ibrahim, KS Abd-Elrahman, S El Mestikawy, SSG Ferguson
    Molecular pharmacology 98 (4), 314-327 2020
    Citations: 18

  • Vascular smooth muscle-specific EP4 receptor deletion in mice exacerbates angiotensin II-induced renal injury
    JF Thibodeau, CE Holterman, Y He, A Carter, GO Cron, NC Boisvert, ...
    Antioxidants & redox signaling 25 (12), 642-656 2016
    Citations: 18