Network


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

Hotspot


Dive into the research topics where Azeb Tadesse Argaw is active.

Publication


Featured researches published by Azeb Tadesse Argaw.


Proceedings of the National Academy of Sciences of the United States of America | 2009

VEGF-mediated disruption of endothelial CLN-5 promotes blood-brain barrier breakdown

Azeb Tadesse Argaw; Blake T. Gurfein; Yueting Zhang; Andleeb Zameer; Gareth R. John

Breakdown of the blood-brain barrier (BBB) is an early and significant event in CNS inflammation. Astrocyte-derived VEGF-A has been implicated in this response, but the underlying mechanisms remain unresolved. Here, we identify the endothelial transmembrane tight junction proteins claudin-5 (CLN-5) and occludin (OCLN) as targets of VEGF-A action. Down-regulation of CLN-5 and OCLN accompanied up-regulation of VEGF-A and correlated with BBB breakdown in experimental autoimmune encephalomyelitis, an animal model of CNS inflammatory disease. In cultures of brain microvascular endothelial cells, VEGF-A specifically down-regulated CLN-5 and OCLN protein and mRNA. In mouse cerebral cortex, microinjection of VEGF-A disrupted CLN-5 and OCLN and induced loss of barrier function. Importantly, functional studies revealed that expression of recombinant CLN-5 protected brain microvascular endothelial cell cultures from a VEGF-induced increase in paracellular permeability, whereas recombinant OCLN expressed under the same promoter was not protective. Previous studies have shown CLN-5 to be a key determinant of trans-endothelial resistance at the BBB. Our findings suggest that its down-regulation by VEGF-A constitutes a significant mechanism in BBB breakdown.


Journal of Clinical Investigation | 2012

Astrocyte-derived VEGF-A drives blood-brain barrier disruption in CNS inflammatory disease

Azeb Tadesse Argaw; Linnea Asp; Jingya Zhang; Kristina Navrazhina; Trinh Pham; John N. Mariani; Sean Mahase; Dipankar J. Dutta; Jeremy Seto; Elisabeth G. Kramer; Napoleone Ferrara; Michael V. Sofroniew; Gareth R. John

In inflammatory CNS conditions such as multiple sclerosis (MS), current options to treat clinical relapse are limited, and more selective agents are needed. Disruption of the blood-brain barrier (BBB) is an early feature of lesion formation that correlates with clinical exacerbation, leading to edema, excitotoxicity, and entry of serum proteins and inflammatory cells. Here, we identify astrocytic expression of VEGF-A as a key driver of BBB permeability in mice. Inactivation of astrocytic Vegfa expression reduced BBB breakdown, decreased lymphocyte infiltration and neuropathology in inflammatory and demyelinating lesions, and reduced paralysis in a mouse model of MS. Knockdown studies in CNS endothelium indicated activation of the downstream effector eNOS as the principal mechanism underlying the effects of VEGF-A on the BBB. Systemic administration of the selective eNOS inhibitor cavtratin in mice abrogated VEGF-A-induced BBB disruption and pathology and protected against neurologic deficit in the MS model system. Collectively, these data identify blockade of VEGF-A signaling as a protective strategy to treat inflammatory CNS disease.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Notch1 signaling plays a role in regulating precursor differentiation during CNS remyelination

Yueting Zhang; Azeb Tadesse Argaw; Blake T. Gurfein; Andleeb Zameer; Brian J. Snyder; Changhui Ge; Q. Richard Lu; David H. Rowitch; Cedric S. Raine; Celia F. Brosnan; Gareth R. John

In the developing CNS, Notch1 and its ligand, Jagged1, regulate oligodendrocyte differentiation and myelin formation, but their role in repair of demyelinating lesions in diseases such as multiple sclerosis remains unresolved. To address this question, we generated a mouse model in which we targeted Notch1 inactivation to oligodendrocyte progenitor cells (OPCs) using Olig1Cre and a floxed Notch1 allele, Notch112f. During CNS development, OPC differentiation was potentiated in Olig1Cre:Notch112f/12f mice. Importantly, in adults, remyelination of demyelinating lesions was also accelerated, at the expense of proliferation within the progenitor population. Experiments in vitro confirmed that Notch1 signaling was permissive for OPC expansion but inhibited differentiation and myelin formation. These studies also revealed that astrocytes exposed to TGF-β1 restricted OPC maturation via Jagged1-Notch1 signaling. These data suggest that Notch1 signaling is one of the mechanisms regulating OPC differentiation during CNS remyelination. Thus, Notch1 may represent a potential therapeutical avenue for lesion repair in demyelinating disease.


Journal of Immunology | 2009

IL-11 Regulates Autoimmune Demyelination

Blake T. Gurfein; Yueting Zhang; Carolina B. López; Azeb Tadesse Argaw; Andleeb Zameer; Thomas M. Moran; Gareth R. John

Current therapies for the autoimmune demyelinating disease multiple sclerosis (MS) target inflammation, but do not directly address neuroprotection or lesion repair. Cytokines of the gp130 family regulate survival and differentiation of both neural and immune cells, and we recently identified expression of the family member IL-11 in active MS plaques. In this study, we show that IL-11 regulates the clinical course and neuropathology of experimental autoimmune encephalomyelitis, a demyelinating model that mimics many of the clinical and pathologic features of MS. Importantly, the effects of IL-11 are achieved via a combination of immunoregulation and direct neuroprotection. IL-11R-α-null (IL-11Rα−/−) mice displayed a significant increase in clinical severity and neuropathology of experimental autoimmune encephalomyelitis compared with wild-type littermates. Inflammation, demyelination, and oligodendrocyte and neuronal loss were all exacerbated in IL-11Ra−/− animals. Conversely, wild-type mice treated with IL-11 displayed milder clinical signs and neuropathology than vehicle-treated controls. In cocultures of murine myelin oligodendrocyte glycoprotein35–55-specific CD4+ T lymphocytes and CD11c+ APCs, IL-11 treatment resulted in a significant decrease in T cell-derived effector cytokine production. This effect was generated via modulation of CD11c+ APC-mediated lymphocyte activation, and was associated with a decrease in the size of the CD11c+ cell population. Conversely, IL-11 strongly reduced apoptosis and potentiated mitosis in primary cultures of mouse oligodendrocyte progenitors. Collectively, these data reveal that IL-11 regulates inflammatory demyelination via a unique combination of immunoregulation and neuroprotection. IL-11 signaling may represent a therapeutic avenue to restrict CNS inflammation and potentiate oligodendrocyte survival in autoimmune demyelinating disease.


Brain | 2015

Astrocytic TYMP and VEGFA drive blood-brain barrier opening in inflammatory central nervous system lesions.

Candice Chapouly; Azeb Tadesse Argaw; Sam Horng; Kamilah Castro; Jingya Zhang; Linnea Asp; Hannah Loo; Benjamin M. Laitman; John N. Mariani; Rebecca Straus Farber; Elena Zaslavsky; German Nudelman; Cedric S. Raine; Gareth R. John

In inflammatory central nervous system conditions such as multiple sclerosis, breakdown of the blood-brain barrier is a key event in lesion pathogenesis, predisposing to oedema, excitotoxicity, and ingress of plasma proteins and inflammatory cells. Recently, we showed that reactive astrocytes drive blood-brain barrier opening, via production of vascular endothelial growth factor A (VEGFA). Here, we now identify thymidine phosphorylase (TYMP; previously known as endothelial cell growth factor 1, ECGF1) as a second key astrocyte-derived permeability factor, which interacts with VEGFA to induce blood-brain barrier disruption. The two are co-induced NFκB1-dependently in human astrocytes by the cytokine interleukin 1 beta (IL1B), and inactivation of Vegfa in vivo potentiates TYMP induction. In human central nervous system microvascular endothelial cells, VEGFA and the TYMP product 2-deoxy-d-ribose cooperatively repress tight junction proteins, driving permeability. Notably, this response represents part of a wider pattern of endothelial plasticity: 2-deoxy-d-ribose and VEGFA produce transcriptional programs encompassing angiogenic and permeability genes, and together regulate a third unique cohort. Functionally, each promotes proliferation and viability, and they cooperatively drive motility and angiogenesis. Importantly, introduction of either into mouse cortex promotes blood-brain barrier breakdown, and together they induce severe barrier disruption. In the multiple sclerosis model experimental autoimmune encephalitis, TYMP and VEGFA co-localize to reactive astrocytes, and correlate with blood-brain barrier permeability. Critically, blockade of either reduces neurologic deficit, blood-brain barrier disruption and pathology, and inhibiting both in combination enhances tissue preservation. Suggesting importance in human disease, TYMP and VEGFA both localize to reactive astrocytes in multiple sclerosis lesion samples. Collectively, these data identify TYMP as an astrocyte-derived permeability factor, and suggest TYMP and VEGFA together promote blood-brain barrier breakdown.


Glia | 2010

TGFβ1 induces Jagged1 expression in astrocytes via ALK5 and Smad3 and regulates the balance between oligodendrocyte progenitor proliferation and differentiation

Yueting Zhang; Jingya Zhang; Kristina Navrazhina; Azeb Tadesse Argaw; Andleeb Zameer; Blake T. Gurfein; Celia F. Brosnan; Gareth R. John

Notch1 receptor signaling regulates oligodendrocyte progenitor differentiation and myelin formation in development, and during remyelination in the adult CNS. In active multiple sclerosis lesions, Notch1 localizes to oligodendrocyte lineage cells, and its ligand Jagged1 is expressed by reactive astrocytes. Here, we examined induction of Jagged1 in human astrocytes, and its impact on oligodendrocyte differentiation. In human astrocyte cultures, the cytokine TGFβ1 induced Jagged1 expression and blockade of the TGFβ1 receptor kinase ALK5 abrogated Jagged1 induction. TGFβ2 and β3 had similar effects, but induction was not observed in response to the TGFβ family member activin A or other cytokines. Downstream, TGFβ1 activated Smad‐dependent signaling, and Smad‐independent pathways that included PI3 kinase, p38, and JNK MAP kinase, but only inhibition of the Smad‐dependent pathway blocked Jagged1 expression. SiRNA inhibition of Smad3 downregulated induction of Jagged1, and this was potentiated by Smad2 siRNA. Purified oligodendrocyte progenitor cells (OPCs) nucleofected with Notch1 intracellular signaling domain displayed a shift towards proliferation at the expense of differentiation, demonstrating functional relevance of Notch1 signaling in OPCs. Furthermore, human OPCs plated onto Jagged1‐expressing astrocytes exhibited restricted differentiation. Collectively, these data illustrate the mechanisms underlying Jagged1 induction in human astrocytes, and suggest that TGFβ1‐induced activation of Jagged1‐Notch1 signaling may impact the size and differentiation of the OPC pool in the human CNS.


Journal of Clinical Investigation | 2017

Astrocytic tight junctions control inflammatory CNS lesion pathogenesis

Sam Horng; Anthony Therattil; Sarah Moyon; Alexandra Gordon; Karla Kim; Azeb Tadesse Argaw; Yuko Hara; John N. Mariani; Setsu Sawai; Per Flodby; Edward D. Crandall; Zea Borok; Michael V. Sofroniew; Candice Chapouly; Gareth R. John

Lesions and neurologic disability in inflammatory CNS diseases such as multiple sclerosis (MS) result from the translocation of leukocytes and humoral factors from the vasculature, first across the endothelial blood-brain barrier (BBB) and then across the astrocytic glia limitans (GL). Factors secreted by reactive astrocytes open the BBB by disrupting endothelial tight junctions (TJs), but the mechanisms that control access across the GL are unknown. Here, we report that in inflammatory lesions, a second barrier composed of reactive astrocyte TJs of claudin 1 (CLDN1), CLDN4, and junctional adhesion molecule A (JAM-A) subunits is induced at the GL. In a human coculture model, CLDN4-deficient astrocytes were unable to control lymphocyte segregation. In models of CNS inflammation and MS, mice with astrocyte-specific Cldn4 deletion displayed exacerbated leukocyte and humoral infiltration, neuropathology, motor disability, and mortality. These findings identify a second inducible barrier to CNS entry at the GL. This barrier may be therapeutically targetable in inflammatory CNS disease.


Journal of Immunology | 2011

Proapoptotic and Antiapoptotic Actions of Stat1 versus Stat3 Underlie Neuroprotective and Immunoregulatory Functions of IL-11

Jingya Zhang; Yueting Zhang; Dipankar J. Dutta; Azeb Tadesse Argaw; Virginie Bonnamain; Jeremy Seto; David A. Braun; Andleeb Zameer; Fernand Hayot; Carolina B. López; Cedric S. Raine; Gareth R. John

Current therapies for multiple sclerosis target inflammation but do not directly address oligodendrocyte protection or myelin repair. The gp130 family cytokines ciliary neurotrophic factor, leukemia inhibitory factor, and IL-11 have been identified as oligodendrocyte growth factors, and IL-11 is also strongly immunoregulatory, but their underlying mechanisms of action are incompletely characterized. In this study, we demonstrate that these effects of IL-11 are mediated via differential regulation of apoptosis in oligodendrocytes versus Ag-presenting dendritic cells (DCs), and are dependent on lineage-specific activity of the transcription factors Stat1 versus Stat3. Focal demyelinating lesions induced in cerebral cortices of IL-11Rα−/− mice using stereotactic microinjection of lysolecithin were larger than in controls, and remyelination was delayed. In IL-11Rα−/− mice, lesions displayed extensive oligodendrocyte loss and axonal transection, and increased infiltration by inflammatory cells including CD11c+ DCs, CD3+ lymphocytes, and CD11b+ phagocytes. In oligodendrocyte progenitor cell (OPC) cultures, IL-11 restricted caspase 9 activation and apoptosis, and it increased myelination in OPC-neuron cocultures. Importantly, siRNA inhibition of Stat1 enhanced the antiapoptotic effects of IL-11 on OPCs, but IL-11 induced apoptosis in the presence of Stat3 silencing. In contrast, IL-11 augmented caspase activation and apoptosis in cultures of CD11c+ DCs, but not in CD11b+ or CD3+ cells. Inhibition of Stat3 exacerbated the proapoptotic effects of IL-11 on DCs, whereas they were ablated in Stat1−/− cultures. Collectively, these findings reveal novel mechanisms underlying the actions of a neuroprotective and immunoregulatory member of the gp130 cytokine family, suggesting avenues to enhance oligodendrocyte viability and restrict CNS inflammation in multiple sclerosis.


Development | 2014

Combinatorial actions of Tgfβ and Activin ligands promote oligodendrocyte development and CNS myelination.

Dipankar J. Dutta; Andleeb Zameer; John N. Mariani; Jingya Zhang; Linnea Asp; Jimmy Huynh; Sean Mahase; Benjamin M. Laitman; Azeb Tadesse Argaw; Nesanet Mitiku; Mateusz Urbanski; Patrizia Casaccia; Fernand Hayot; Erwin P. Bottinger; Chester W. Brown; Gareth R. John

In the embryonic CNS, development of myelin-forming oligodendrocytes is limited by bone morphogenetic proteins, which constitute one arm of the transforming growth factor-β (Tgfβ) family and signal canonically via Smads 1/5/8. Tgfβ ligands and Activins comprise the other arm and signal via Smads 2/3, but their roles in oligodendrocyte development are incompletely characterized. Here, we report that Tgfβ ligands and activin B (ActB) act in concert in the mammalian spinal cord to promote oligodendrocyte generation and myelination. In mouse neural tube, newly specified oligodendrocyte progenitors (OLPs) are first exposed to Tgfβ ligands in isolation, then later in combination with ActB during maturation. In primary OLP cultures, Tgfβ1 and ActB differentially activate canonical Smad3 and non-canonical MAP kinase signaling. Both ligands enhance viability, and Tgfβ1 promotes proliferation while ActB supports maturation. Importantly, co-treatment strongly activates both signaling pathways, producing an additive effect on viability and enhancing both proliferation and differentiation such that mature oligodendrocyte numbers are substantially increased. Co-treatment promotes myelination in OLP-neuron co-cultures, and maturing oligodendrocytes in spinal cord white matter display strong Smad3 and MAP kinase activation. In spinal cords of ActB-deficient Inhbb−/− embryos, apoptosis in the oligodendrocyte lineage is increased and OLP numbers transiently reduced, but numbers, maturation and myelination recover during the first postnatal week. Smad3−/− mice display a more severe phenotype, including diminished viability and proliferation, persistently reduced mature and immature cell numbers, and delayed myelination. Collectively, these findings suggest that, in mammalian spinal cord, Tgfβ ligands and ActB together support oligodendrocyte development and myelin formation.


FEBS Letters | 2011

Promoting myelin repair and return of function in multiple sclerosis

Jingya Zhang; Elisabeth G. Kramer; Linnea Asp; Dipankar J. Dutta; Kristina Navrazhina; Trinh Pham; John N. Mariani; Azeb Tadesse Argaw; Gareth R. John

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. Conduction block in demyelinated axons underlies early neurological symptoms, but axonal transection and neuronal loss are believed to be responsible for more permanent chronic deficits. Several therapies are approved for treatment of relapsing‐remitting MS, all of which are immunoregulatory and clinically proven to reduce the rate of lesion formation and exacerbation. However, existing approaches are only partially effective in preventing the onset of disability in MS patients, and novel treatments to protect myelin‐producing oligodendrocytes and enhance myelin repair may improve long‐term outcomes. Studies in vivo in genetically modified mice have assisted in the characterization of mechanisms underlying the generation of neuropathology in MS patients, and have identified potential avenues for oligodendrocyte protection and myelin repair. However, no treatments are yet approved that target these areas directly, and in addition, the relationship between demyelination and axonal transection in the lesions of the disease remains unclear. Here, we review translational research targeting oligodendrocyte protection and myelin repair in models of autoimmune demyelination, and their potential relevance as therapies in MS.

Collaboration


Dive into the Azeb Tadesse Argaw's collaboration.

Top Co-Authors

Avatar

Gareth R. John

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jingya Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

John N. Mariani

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Linnea Asp

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Andleeb Zameer

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Dipankar J. Dutta

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Benjamin M. Laitman

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Sean Mahase

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Fernand Hayot

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yueting Zhang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge