Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Manzoor A. Bhat is active.

Publication


Featured researches published by Manzoor A. Bhat.


Neuron | 2001

Axon-Glia Interactions and the Domain Organization of Myelinated Axons Requires Neurexin IV/Caspr/Paranodin

Manzoor A. Bhat; Jose C. Rios; Yue Lu; German P. Garcia-Fresco; William Ching; Mary St. Martin; Jingjun Li; Steven Einheber; Mitchell Chesler; Jack Rosenbluth; James L. Salzer; Hugo J. Bellen

Myelinated fibers are organized into distinct domains that are necessary for saltatory conduction. These domains include the nodes of Ranvier and the flanking paranodal regions where glial cells closely appose and form specialized septate-like junctions with axons. These junctions contain a Drosophila Neurexin IV-related protein, Caspr/Paranodin (NCP1). Mice that lack NCP1 exhibit tremor, ataxia, and significant motor paresis. In the absence of NCP1, normal paranodal junctions fail to form, and the organization of the paranodal loops is disrupted. Contactin is undetectable in the paranodes, and K(+) channels are displaced from the juxtaparanodal into the paranodal domains. Loss of NCP1 also results in a severe decrease in peripheral nerve conduction velocity. These results show a critical role for NCP1 in the delineation of specific axonal domains and the axon-glia interactions required for normal saltatory conduction.


Cell | 1996

A Drosophila Neurexin Is Required for Septate Junction and Blood-Nerve Barrier Formation and Function

Stefan Baumgartner; J. Troy Littleton; Kendal Broadie; Manzoor A. Bhat; Ruth Harbecke; Judith A. Lengyel; Ruth Chiquet-Ehrismann; Andreas Prokop; Hugo J. Bellen

Septate and tight junctions are thought to seal neighboring cells together and to function as barriers between epithelial cells. We have characterized a novel member of the neurexin family, Neurexin IV (NRX), which is localized to septate junctions (SJs) of epithelial and glial cells. NRX is a transmembrane protein with a cytoplasmic domain homologous to glycophorin C, a protein required for anchoring protein 4.1 in the red blood cell. Absence of NRX results in mislocalization of Coracle, a Drosophila protein 4.1 homolog, at SJs and causes dorsal closure defects similar to those observed in coracle mutants. nrx mutant embryos are paralyzed, and electrophysiological studies indicate that the lack of NRX in glial-glial SJs causes a breakdown of the blood-brain barrier. Electron microscopy demonstrates that nrx mutants lack the ladder-like intercellular septa characteristic of pleated SJs (pSJs). These studies identify NRX as the first transmembrane protein of SJ and demonstrate a requirement for NRX in the formation of septate-junction septa and intercellular barriers.


Cell | 1996

Chromatid segregation at anaphase requires the barren product, a novel chromosome-associated protein that interacts with topoisomerase II

Manzoor A. Bhat; Alastair Valentine Philp; David M. Glover; Hugo J. Bellen

We have isolated a Drosophila gene, barren (barr), required for sister-chromatid segregation in mitosis. barr encodes a novel protein that is present in proliferating cells and has homologs in yeast and human. Mitotic defects in barr embryos become apparent during cycle 16, resulting in a loss of PNS and CNS neurons. Centromeres move apart at the metaphase-anaphase transition and Cyclin B is degraded, but sister chromatids remain connected, resulting in chromatin bridging. This phenotype is similar to that described in TOP2 mutants in yeast. Barren protein localizes to chromatin throughout mitosis. Colocalization and biochemical experiments indicate that Barren associates with Topoisomerase II throughout mitosis and alters the activity of Topoisomerase II. We propose that this association is required for proper chromosomal segregation by facilitating the decatenation of chromatids at anaphase.


Cell | 1999

Discs Lost, a Novel Multi-PDZ Domain Protein, Establishes and Maintains Epithelial Polarity

Manzoor A. Bhat; Shayan A. Izaddoost; Yue Lu; Kyung-Ok Cho; Kwang Wook Choi; Hugo J. Bellen

Polarization of epithelial cells depends on a hierarchical process whereby specific membrane-associated proteins become targeted to specialized membrane domains. Here, we describe a novel Drosophila protein, Discs Lost (DLT), that plays a crucial role in the polarization of embryonic epithelia during cellular blastoderm formation. At subsequent stages of development, DLT interacts with the apical determinant Crumbs (CRB) and the laterally localized protein Neurexin IV (NRX IV). Mutations in dlt or double-stranded RNA interference lead to aberrant localization of CRB and NRX IV and cause a concomitant loss of epithelial cell polarity. Hence, DLT is required to establish and maintain cell polarity and participates in different molecular complexes that define apical and lateral membrane domains.


Nature | 2002

Drosophila Crumbs is a positional cue in photoreceptor adherens junctions and rhabdomeres

Shayan A. Izaddoost; Sang Chul Nam; Manzoor A. Bhat; Hugo J. Bellen; Kwang Wook Choi

Drosophila Crumbs (Crb) is required for apical–basal polarity and is an apical determinant in embryonic epithelia. Here, we describe properties of Crb that control the position and integrity of the photoreceptor adherens junction and photosensitive organ, or rhabdomere. In contrast to normal photoreceptor adherens junctions and rhabdomeres, which span the depth of the retina, adherens junctions and rhabdomeres of Crb-deficient photoreceptors initially accumulate at the top of the retina and fail to maintain their integrity as they stretch to the retinal floor. We show that Crb controls localization of the adherens junction through its intracellular domain containing a putative binding site for a protein 4.1 superfamily protein (FERM). Although loss of Crb or overexpression of the FERM binding domain causes mislocalization of adherens junctions, they do not result in a significant loss of photoreceptor polarity. Mutations in CRB1, a human homologue of crb, are associated with photoreceptor degeneration in retinitis pigmentosa 12 (RP12) and Leber congenital amaurosis (LCA). The intracellular domain of CRB1 behaves similarly to its Drosophila counterpart when overexpressed in the fly eye. Our studies may provide clues for mechanisms of photoreceptor degeneration in RP12 and LCA.


The EMBO Journal | 1998

ROP, the Drosophila Sec1 homolog, interacts with syntaxin and regulates neurotransmitter release in a dosage-dependent manner

Mark N. Wu; J. Troy Littleton; Manzoor A. Bhat; Andreas Prokop; Hugo J. Bellen

The Sec1 family of proteins is thought to function in both non‐neuronal and neuronal secretion, although the precise role of this protein family has not been defined. Here, we study the function of ROP, the Drosophila Sec1 homolog, in neurotransmitter release. Electrophysiological analyses of transgenic lines overexpressing ROP and syntaxin, a presynaptic membrane protein, indicate that ROP interacts with syntaxin in vivo. Characterization of four point mutations in ROP shows that they fall into two phenotypic classes. Two mutations cause a dramatic reduction in both evoked and spontaneous neurotransmitter release. In contrast, the other two mutations reveal an increase in evoked neurotransmission. Our data further show that neurotransmission is highly sensitive to the levels of ROP function. Studies on heterozygote animals indicate that half the amount of wild‐type ROP results in a dramatic decrease in evoked and spontaneous exocytosis. Taken together, these results suggest that ROP interacts with syntaxin in vivo and is a rate‐limiting regulator of exocytosis that performs both positive and inhibitory functions in neurotransmission.


Development | 2004

Drosophila contactin, a homolog of vertebrate contactin, is required for septate junction organization and paracellular barrier function

Catherine Faivre-Sarrailh; Swati Banerjee; Jingjun Li; Michael Hortsch; Monique Laval; Manzoor A. Bhat

Septate junctions (SJs) in epithelial and neuronal cells play an important role in the formation and maintenance of charge and size selective barriers. They form the basis for the ensheathment of nerve fibers in Drosophila and for the attachment of myelin loops to axonal surface in vertebrates. The cell-adhesion molecules NRX IV/Caspr/Paranodin (NCP1), contactin and Neurofascin-155 (NF-155) are all present at the vertebrate axo-glial SJs. Mutational analyses have shown that vertebrate NCP1 and its Drosophila homolog, Neurexin IV (NRX IV) are required for the formation of SJs. In this study, we report the genetic, molecular and biochemical characterization of the Drosophila homolog of vertebrate contactin, CONT. Ultrastructural and dye-exclusion analyses of Cont mutant embryos show that CONT is required for organization of SJs and paracellular barrier function. We show that CONT, Neuroglian (NRG) (Drosophila homolog of NF-155) and NRX IV are interdependent for their SJ localization and these proteins form a tripartite complex. Hence, our data provide evidence that the organization of SJs is dependent on the interactions between these highly conserved cell-adhesion molecules.


Trends in Neurosciences | 1998

Neurexin IV, caspr and paranodin—novel members of the neurexin family: encounters of axons and glia

Hugo J. Bellen; Yue Lu; Robert B. Beckstead; Manzoor A. Bhat

Axonal insulation is of key importance for the proper propagation of action potentials. In Drosophila and other invertebrates, it has recently been demonstrated that septate junctions play an essential role in axonal insulation or blood-brain-barrier formation. Neurexin IV, a molecular component of Drosophila septate junctions, has been shown to be essential for axonal insulation in the PNS in embryos and larvae. Interestingly, a vertebrate homolog of Neurexin IV, caspr--also named paranodin--has been shown to localize to septate-like junctional structures. These vertebrate junctions are localized to the paranodal region of the nodes of Ranvier, between axons and Schwann cells. Caspr/paranodin might play an important role in barrier formation, and link neuronal membrane components with the axonal cytoskeletal network.


Journal of Neuroscience Research | 2009

Spatiotemporal Ablation of Myelinating Glia-Specific Neurofascin (NfascNF155) in Mice Reveals Gradual Loss of Paranodal Axoglial Junctions and Concomitant Disorganization of Axonal Domains

Anilkumar M. Pillai; Courtney Thaxton; Alaine L. Pribisko; JrGang Cheng; Jeffrey L. Dupree; Manzoor A. Bhat

The evolutionary demand for rapid nerve impulse conduction led to the process of myelination‐dependent organization of axons into distinct molecular domains. These domains include the node of Ranvier flanked by highly specialized paranodal domains where myelin loops and axolemma orchestrate the axoglial septate junctions. These junctions are formed by interactions between a glial isoform of neurofascin (NfascNF155) and axonal Caspr and Cont. Here we report the generation of myelinating glia‐specific NfascNF155 null mouse mutants. These mice exhibit severe ataxia, motor paresis, and death before the third postnatal week. In the absence of glial NfascNF155, paranodal axoglial junctions fail to form, axonal domains fail to segregate, and myelinated axons undergo degeneration. Electrophysiological measurements of peripheral nerves from NfascNF155 mutants revealed dramatic reductions in nerve conduction velocities. By using inducible PLP‐CreER recombinase to ablate NfascNF155 in adult myelinating glia, we demonstrate that paranodal axoglial junctions disorganize gradually as the levels of NfascNF155 protein at the paranodes begin to drop. This coincides with the loss of the paranodal region and concomitant disorganization of the axonal domains. Our results provide the first direct evidence that the maintenance of axonal domains requires the fence function of the paranodal axoglial junctions. Together, our studies establish a central role for paranodal axoglial junctions in both the organization and the maintenance of axonal domains in myelinated axons.


Neuron | 2007

Crucial Role of Drosophila Neurexin in Proper Active Zone Apposition to Postsynaptic Densities, Synaptic Growth, and Synaptic Transmission

Jingjun Li; James A. Ashley; Vivian Budnik; Manzoor A. Bhat

Neurexins have been proposed to function as major mediators of the coordinated pre- and postsynaptic apposition. However, key evidence for this role in vivo has been lacking, particularly due to gene redundancy. Here, we have obtained null mutations in the single Drosophila neurexin gene (dnrx). dnrx loss of function prevents the normal proliferation of synaptic boutons at glutamatergic neuromuscular junctions, while dnrx gain of function in neurons has the opposite effect. DNRX mostly localizes to the active zone of presynaptic terminals. Conspicuously, dnrx null mutants display striking defects in synaptic ultrastructure, with the presence of detachments between pre- and postsynaptic membranes, abnormally long active zones, and increased number of T bars. These abnormalities result in corresponding alterations in synaptic transmission with reduced quantal content. Together, our results provide compelling evidence for an in vivo role of neurexins in the modulation of synaptic architecture and adhesive interactions between pre- and postsynaptic compartments.

Collaboration


Dive into the Manzoor A. Bhat's collaboration.

Top Co-Authors

Avatar

Swati Banerjee

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Hugo J. Bellen

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jeffrey L. Dupree

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Anilkumar M. Pillai

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Anna M. Taylor

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Julia Saifetiarova

University of Texas Health Science Center at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Aurea D. Sousa

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Elizabeth D. Buttermore

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

German P. Garcia-Fresco

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jingjun Li

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge