Network


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

Hotspot


Dive into the research topics where Nithi Asavapanumas is active.

Publication


Featured researches published by Nithi Asavapanumas.


The FASEB Journal | 2014

Greatly improved survival and neuroprotection in aquaporin-4-knockout mice following global cerebral ischemia

Ryuichi Katada; Gökhan Akdemir; Nithi Asavapanumas; Julien Ratelade; Hua Zhang; A. S. Verkman

Aquaporin‐4 (AQP4), the principal water channel in astrocytes, is involved in brain water movement, inflammation, and neuroexcitation. In this study, there was strong neuroprotection in mice lacking AQP4 in a model of global cerebral ischemia produced by transient, bilateral carotid artery occlusion (BCAO). Survival and neurological outcome were greatly improved in the AQP4‐/‐ vs. AQP4+/+ mice after occlusion, with large and robust differences in both outbred (CD1) and inbred (C57bl/6) mouse strains without or with mechanical ventilation. Improved survival was also seen in mice lacking the scaffold protein α‐syntrophin, which manifest reduced astrocyte water permeability secondary to defective AQP4 plasma membrane targeting. Intracranial pressure elevation and brain water accumulation were much reduced in the AQP4‐/‐ vs. AQP4+/+ mice after carotid artery occlusion, as were blood–brain barrier (BBB) disruption and neuronal loss. Brain slices from AQP4‐/‐ mice showed significantly reduced cell swelling and cytotoxicity in response to oxygen–glucose deprivation, compared with slices from AQP4+/+ mice. Our findings suggest that the neuroprotective effect of AQP4 deletion in global cerebral ischemia involves reduced astrocyte swelling and brain water accumulation, resulting in reduced BBB disruption, inflammation, and neuron death. AQP4 water transport inhibition may improve survival and neurological outcome after cardiac arrest and in other conditions associated with global cerebral ischemia.—Katada, R., Akdemir, G., Asavapanumas, N., Ratelade, J., Zhang, H., Verkman, A. S. Greatly improved survival and neuroprotection in aquaporin‐4 knockout mice following global cerebral ischemia. FASEB J. 28, 705–714 (2014). www.fasebj.org


Acta Neuropathologica | 2013

Involvement of antibody-dependent cell-mediated cytotoxicity in inflammatory demyelination in a mouse model of neuromyelitis optica

Julien Ratelade; Nithi Asavapanumas; Alanna M. Ritchie; Scott Wemlinger; Jeffrey L. Bennett; A. S. Verkman

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system that can cause paralysis and blindness. The pathogenesis of NMO involves binding of immunoglobulin G autoantibodies to aquaporin-4 (AQP4) on astrocytes, which is thought to cause complement-dependent cytotoxicity (CDC) and a secondary inflammatory response leading to oligodendrocyte and neuronal damage. Here, we investigate in vivo the role of antibody-dependent cell-mediated cytotoxicity (ADCC) triggered by AQP4 autoantibodies (AQP4-IgG) in the development of NMO pathology. A high-affinity, human recombinant monoclonal AQP4-IgG was mutated in its Fc region to produce ‘NMO superantibodies’ with enhanced CDC and/or ADCC effector functions, without altered AQP4 binding. Pathological effects of these antibodies were studied in a mouse model of NMO produced by intracerebral injection of AQP4-IgG and human complement. The original (non-mutated) antibody produced large NMO lesions in this model, with loss of AQP4 and GFAP immunoreactivity, inflammation and demyelination, as did a mutated antibody with enhanced CDC and ADCC effector functions. As anticipated, a mutated AQP4-IgG lacking CDC, but having tenfold enhanced ADCC, produced little pathology. However, unexpectedly, a mutated antibody with ninefold enhanced CDC, but lacking ADCC, produced much less pathology than the original AQP4-IgG. Also, pathology was greatly reduced following administration of AQP4-IgG and complement to mice lacking the FcγIII receptor involved in effector cell activation during ADCC, and to normal mice injected with an Fcγ receptor blocking antibody. Our results provide evidence for the central involvement of ADCC in NMO pathology and suggest ADCC as a new therapeutic target in NMO.


Molecular Pharmacology | 2013

Therapeutic Cleavage of Anti–Aquaporin-4 Autoantibody in Neuromyelitis Optica by an IgG-Selective Proteinase

Lukmanee Tradtrantip; Nithi Asavapanumas; A. S. Verkman

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system caused by binding of pathogenic IgG autoantibodies (NMO-IgG) to astrocyte water channel aquaporin-4 (AQP4). Astrocyte damage and downstream inflammation require NMO-IgG effector function to initiate complement-dependent cytotoxicity (CDC) and antibody-dependent cell-mediated cytotoxicity (ADCC). Here, we evaluated the potential therapeutic utility of the bacterial enzyme IdeS (IgG-degrading enzyme of Streptococcus pyogenes), which selectively cleaves IgG antibodies to yield Fc and F(ab′)2 fragments. In AQP4-expressing cell cultures, IdeS treatment of monoclonal NMO-IgGs and NMO patient sera abolished CDC and ADCC, even when IdeS was added after NMO-IgG was bound to AQP4. Binding of NMO-IgG to AQP4 was similar to that of the NMO-F(ab′)2 generated by IdeS cleavage. NMO-F(ab′)2 competitively displaced pathogenic NMO-IgG, preventing cytotoxicity, and the Fc fragments generated by IdeS cleavage reduced CDC and ADCC. IdeS efficiently cleaved NMO-IgG in mice in vivo, and greatly reduced NMO lesions in mice administered NMO-IgG and human complement. IgG-selective cleavage by IdeS thus neutralizes NMO-IgG pathogenicity, and yields therapeutic F(ab′)2 and Fc fragments. IdeS treatment, by therapeutic apheresis or direct administration, may be beneficial in NMO.


Neuroscience Letters | 2014

Neuroprotective effect of aquaporin-4 deficiency in a mouse model of severe global cerebral ischemia produced by transient 4-vessel occlusion.

Gökhan Akdemir; Julien Ratelade; Nithi Asavapanumas; A. S. Verkman

Astrocyte water channel aquaporin-4 (AQP4) facilitates water movement across the blood-brain barrier and into injured astrocytes. We previously showed reduced cytotoxic brain edema with improved neurological outcome in AQP4 knockout mice in water intoxication, infection and cerebral ischemia. Here, we established a 4-vessel transient occlusion model to test the hypothesis that AQP4 deficiency in mice could improve neurological outcome following severe global cerebral ischemia as occurs in cardiac arrest/resuscitation. Mice were subjected to 10-min transient bilateral carotid artery occlusion at 24h after bilateral vertebral artery cauterization. Cerebral blood flow was reduced during occlusion by >94% in both AQP4(+/+) and AQP4(-/-) mice. The primary outcome, neurological score, was remarkably better at 3 and 5 days after occlusion in AQP4(-/-) than in AQP4(+/+) mice, and survival was significantly improved as well. Brain water content was increased by 2.8±0.4% in occluded AQP4(+/+) mice, significantly greater than that of 0.3±0.6% in AQP4(-/-) mice. Histological examination and immunofluorescence of hippocampal sections at 5 days showed significantly greater neuronal loss in the CA1 region of hippocampus in AQP4(+/+) than AQP4(-/-) mice. The neuroprotection in mice conferred by AQP4 deletion following severe global cerebral ischemia provides proof-of-concept for therapeutic AQP4 inhibition to improve neurological outcome in cardiac arrest.


Brain Pathology | 2013

Biology of AQP4 and Anti-AQP4 Antibody: Therapeutic Implications for NMO

A. S. Verkman; Puay-Wah Phuan; Nithi Asavapanumas; Lukmanee Tradtrantip

The water channel aquaporin‐4 (AQP4) is the target of the immunoglobulin G autoantibody (AQP4‐IgG) in neuromyelitis optica (NMO). AQP4 is expressed in foot processes of astrocytes throughout the central nervous system, as well as in skeletal muscle and epithelial cells in kidney, lung and gastrointestinal organs. Phenotype analysis of AQP4 knockout mice indicates the involvement of AQP4 in water movement into and out of the brain, astrocyte migration, glial scar formation and neuroexcitatory phenomena. AQP4 monomers form tetramers in membranes, which further aggregate to form supramolecular assemblies called orthogonal arrays of particles. AQP4‐IgG is pathogenic in NMO by a mechanism involving complement‐ and cell‐mediated astrocyte cytotoxicity, which produces an inflammatory response with oligodendrocyte injury and demyelination. AQP4 orthogonal arrays are crucial in NMO pathogenesis, as they increase AQP4‐IgG binding to AQP4 and greatly enhance complement‐dependent cytotoxicity. Novel NMO therapeutics are under development that target AQP4‐IgG or AQP4, including aquaporumab monoclonal antibodies and small molecules that block AQP4‐IgG binding to AQP4, and enzymatic inactivation strategies to neutralize AQP4‐IgG pathogenicity.


Journal of Neuroinflammation | 2014

Experimental mouse model of optic neuritis with inflammatory demyelination produced by passive transfer of neuromyelitis optica-immunoglobulin G.

Nithi Asavapanumas; Julien Ratelade; Marios C. Papadopoulos; Jeffrey L. Bennett; Marc H. Levin; A. S. Verkman

BackgroundAlthough optic neuritis (ON) is a defining feature of neuromyelitis optica (NMO), appropriate animal models of NMO ON are lacking. Most NMO patients are seropositive for immunoglobulin G autoantibodies (NMO-IgG) against the astrocyte water channel aquaporin-4 (AQP4).MethodsSeveral approaches were tested to develop a robust, passive-transfer mouse model of NMO ON, including NMO-IgG and complement delivery by: (i) retrobulbar infusion; (ii) intravitreal injection; (iii) a single intracranial injection near the optic chiasm; and (iv) 3-days continuous intracranial infusion near the optic chiasm.ResultsLittle ON or retinal pathology was seen using approaches (i) to (iii). Using approach (iv), however, optic nerves showed characteristic NMO pathology, with loss of AQP4 and glial fibrillary acidic protein immunoreactivity, granulocyte and macrophage infiltration, deposition of activated complement, demyelination and axonal injury. Even more extensive pathology was created in mice lacking complement inhibitor protein CD59, or using a genetically modified NMO-IgG with enhanced complement effector function, including significant loss of retinal ganglion cells. In control studies, optic nerve pathology was absent in treated AQP4-deficient mice, or in wild-type mice receiving control (non-NMO) IgG and complement.ConclusionPassive transfer of NMO-IgG and complement by continuous infusion near the optic chiasm in mice is sufficient to produce ON with characteristic NMO pathology. The mouse model of NMO ON should be useful in further studies of NMO pathogenesis mechanisms and therapeutics.


PLOS ONE | 2014

Potential Therapeutic Benefit of C1-Esterase Inhibitor in Neuromyelitis Optica Evaluated In Vitro and in an Experimental Rat Model

Lukmanee Tradtrantip; Nithi Asavapanumas; Puay-Wah Phuan; A. S. Verkman

Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system in which binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to astrocytes causes complement-dependent cytotoxicity (CDC) and inflammation resulting in oligodendrocyte and neuronal injury. There is compelling evidence for a central role of complement in NMO pathogenesis. Here, we evaluated the potential of C1-esterase inhibitor (C1-inh) for complement-targeted therapy of NMO. C1-inh is an anti-inflammatory plasma protein with serine protease inhibition activity that has a broad range of biological activities on the contact (kallikrein), coagulation, fibrinolytic and complement systems. C1-inh is approved for therapy of hereditary angioedema (HAE) and has been studied in a small safety trial in acute NMO relapses (NCT 01759602). In vitro assays of NMO-IgG-dependent CDC showed C1-inh inhibition of human and rat complement, but with predicted minimal complement inhibition activity at a dose of 2000 units in humans. Inhibition of complement by C1-inh was potentiated by ∼10-fold by polysulfated macromolecules including heparin and dextran sulfate. In rats, intravenous C1-inh at a dose 30-fold greater than that approved to treat HAE inhibited serum complement activity by <5%, even when supplemented with heparin. Also, high-dose C1-inh did not reduce pathology in a rat model of NMO produced by intracerebral injection of NMO-IgG. Therefore, although C1r and C1s are targets of C1-inh, our in vitro data with human serum and in vivo data in rats suggest that the complement inhibition activity of C1-inh in serum is too low to confer clinical benefit in NMO.


Brain Pathology | 2013

Biology of AQP4 and anti-AQP4 antibody: therapeutic implications

A. S. Verkman; Puay-Wah Phuan; Nithi Asavapanumas; Lukmanee Tradtrantip

The water channel aquaporin‐4 (AQP4) is the target of the immunoglobulin G autoantibody (AQP4‐IgG) in neuromyelitis optica (NMO). AQP4 is expressed in foot processes of astrocytes throughout the central nervous system, as well as in skeletal muscle and epithelial cells in kidney, lung and gastrointestinal organs. Phenotype analysis of AQP4 knockout mice indicates the involvement of AQP4 in water movement into and out of the brain, astrocyte migration, glial scar formation and neuroexcitatory phenomena. AQP4 monomers form tetramers in membranes, which further aggregate to form supramolecular assemblies called orthogonal arrays of particles. AQP4‐IgG is pathogenic in NMO by a mechanism involving complement‐ and cell‐mediated astrocyte cytotoxicity, which produces an inflammatory response with oligodendrocyte injury and demyelination. AQP4 orthogonal arrays are crucial in NMO pathogenesis, as they increase AQP4‐IgG binding to AQP4 and greatly enhance complement‐dependent cytotoxicity. Novel NMO therapeutics are under development that target AQP4‐IgG or AQP4, including aquaporumab monoclonal antibodies and small molecules that block AQP4‐IgG binding to AQP4, and enzymatic inactivation strategies to neutralize AQP4‐IgG pathogenicity.


Brain Pathology | 2013

Biology of AQP4 and Anti-AQP4 Antibody: Therapeutic Implications for NMO: AQP4 and AQP4-IgG in NMO

A. S. Verkman; Puay-Wah Phuan; Nithi Asavapanumas; Lukmanee Tradtrantip

The water channel aquaporin‐4 (AQP4) is the target of the immunoglobulin G autoantibody (AQP4‐IgG) in neuromyelitis optica (NMO). AQP4 is expressed in foot processes of astrocytes throughout the central nervous system, as well as in skeletal muscle and epithelial cells in kidney, lung and gastrointestinal organs. Phenotype analysis of AQP4 knockout mice indicates the involvement of AQP4 in water movement into and out of the brain, astrocyte migration, glial scar formation and neuroexcitatory phenomena. AQP4 monomers form tetramers in membranes, which further aggregate to form supramolecular assemblies called orthogonal arrays of particles. AQP4‐IgG is pathogenic in NMO by a mechanism involving complement‐ and cell‐mediated astrocyte cytotoxicity, which produces an inflammatory response with oligodendrocyte injury and demyelination. AQP4 orthogonal arrays are crucial in NMO pathogenesis, as they increase AQP4‐IgG binding to AQP4 and greatly enhance complement‐dependent cytotoxicity. Novel NMO therapeutics are under development that target AQP4‐IgG or AQP4, including aquaporumab monoclonal antibodies and small molecules that block AQP4‐IgG binding to AQP4, and enzymatic inactivation strategies to neutralize AQP4‐IgG pathogenicity.


Acta Neuropathologica | 2013

C1q-targeted monoclonal antibody prevents complement-dependent cytotoxicity and neuropathology in in vitro and mouse models of neuromyelitis optica.

Puay-Wah Phuan; Hua Zhang; Nithi Asavapanumas; Michael Leviten; Arnon Rosenthal; Lukmanee Tradtrantip; A. S. Verkman

Collaboration


Dive into the Nithi Asavapanumas's collaboration.

Top Co-Authors

Avatar

A. S. Verkman

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Puay-Wah Phuan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hua Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Jeffrey L. Bennett

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Alanna M. Ritchie

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marc H. Levin

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge