Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Neuroinflammation ; 16(1): 57, 2019 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-30851734

RESUMO

BACKGROUND: Neuromyelitis optica spectrum disorder (herein called NMO) is an inflammatory demyelinating disease that can be initiated by binding of immunoglobulin G autoantibodies (AQP4-IgG) to aquaporin-4 on astrocytes, causing complement-dependent cytotoxicity (CDC) and downstream inflammation. The increased NMO pathology in rodents deficient in complement regulator protein CD59 following passive transfer of AQP4-IgG has suggested the potential therapeutic utility of increasing the expression of complement regulator proteins. METHODS: A cell-based ELISA was developed to screen for pharmacological upregulators of endogenous CD55 and CD59 in a human astrocyte cell line. A statin identified from the screen was characterized in cell culture models and rodents for its action on complement regulator protein expression and its efficacy in models of seropositive NMO. RESULTS: Screening of ~ 11,500 approved and investigational drugs and nutraceuticals identified transcriptional upregulators of CD55 but not of CD59. Several statins, including atorvastatin, simvastatin, lovastatin, and fluvastatin, increased CD55 protein expression in astrocytes, including primary cultures, by three- to four-fold at 24 h, conferring significant protection against AQP4-IgG-induced CDC. Mechanistic studies revealed that CD55 upregulation involves inhibition of the geranylgeranyl transferase pathway rather than inhibition of cholesterol biosynthesis. Oral atorvastatin at 10-20 mg/kg/day for 3 days strongly increased CD55 immunofluorescence in mouse brain and spinal cord and reduced NMO pathology following intracerebral AQP4-IgG injection. CONCLUSION: Atorvastatin or other statins may thus have therapeutic benefit in AQP4-IgG seropositive NMO by increasing CD55 expression, in addition to their previously described anti-inflammatory and immunomodulatory actions.


Assuntos
Aquaporina 4/imunologia , Astrócitos/metabolismo , Antígenos CD55/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Imunoglobulina G/administração & dosagem , Neuromielite Óptica/tratamento farmacológico , Regulação para Cima/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Linhagem Celular Transformada , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/química , Camundongos , Neuromielite Óptica/metabolismo , Neuromielite Óptica/patologia , RNA Mensageiro/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Ativação Transcricional/efeitos dos fármacos , Receptor fas/metabolismo
2.
FASEB J ; 31(2): 751-760, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27871064

RESUMO

Secretory diarrheas caused by bacterial enterotoxins, including cholera and traveler's diarrhea, remain a major global health problem. Inappropriate activation of the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel occurs in these diarrheas. We previously reported that the benzopyrimido-pyrrolo-oxazinedione (R)-BPO-27 inhibits CFTR chloride conductance with low-nanomolar potency. Here, we demonstrate using experimental mouse models and human enterocyte cultures the potential utility of (R)-BPO-27 for treatment of secretory diarrheas caused by cholera and Escherichia coli enterotoxins. (R)-BPO-27 fully blocked CFTR chloride conductance in epithelial cell cultures and intestine after cAMP agonists, cholera toxin, or heat-stable enterotoxin of E. coli (STa toxin), with IC50 down to ∼5 nM. (R)-BPO-27 prevented cholera toxin and STa toxin-induced fluid accumulation in small intestinal loops, with IC50 down to 0.1 mg/kg. (R)-BPO-27 did not impair intestinal fluid absorption or inhibit other major intestinal transporters. Pharmacokinetics in mice showed >90% oral bioavailability with sustained therapeutic serum levels for >4 h without the significant toxicity seen with 7-d administration at 5 mg/kg/d. As evidence to support efficacy in human diarrheas, (R)-BPO-27 blocked fluid secretion in primary cultures of enteroids from human small intestine and anion current in enteroid monolayers. These studies support the potential utility of (R)-BPO-27 for therapy of CFTR-mediated secretory diarrheas.-Cil, O., Phuan, P.-W., Gillespie, A. M., Lee, S., Tradtrantip, L., Yin, J., Tse, M., Zachos, N. C., Lin, R., Donowitz, M., Verkman, A. S. Benzopyrimido-pyrrolo-oxazine-dione CFTR inhibitor (R)-BPO-27 for antisecretory therapy of diarrheas caused by bacterial enterotoxins.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Diarreia/induzido quimicamente , Diarreia/tratamento farmacológico , Oxazinas/farmacologia , Pirimidinonas/farmacologia , Pirróis/farmacologia , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Intestinos/efeitos dos fármacos , Camundongos , Estrutura Molecular , Oxazinas/síntese química , Pirimidinonas/síntese química , Pirróis/síntese química
3.
Acta Neuropathol ; 134(1): 35-44, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28567523

RESUMO

Neuromyelitis optica spectrum disorder (herein called NMO) is an autoimmune inflammatory disease of the central nervous system in which immunoglobulin G antibodies against astrocyte water channel aquaporin-4 (AQP4-IgG) cause demyelination and neurological deficit. Injury to oligodendrocytes, which do not express AQP4, links the initiating pathogenic event of AQP4-IgG binding to astrocyte AQP4 to demyelination. Here, we report evidence for a complement 'bystander mechanism' to account for early oligodendrocyte injury in NMO in which activated, soluble complement proteins following AQP4-IgG binding to astrocyte AQP4 result in deposition of the complement membrane attack complex (MAC) on nearby oligodendrocytes. Primary cocultures of rat astrocytes and mature oligodendrocytes exposed to AQP4-IgG and complement showed early death of oligodendrocytes in close contact with astrocytes, which was not seen in pure oligodendrocyte cultures, in cocultures exposed to AQP4-IgG and C6-depleted serum, or when astrocytes were damaged by a complement-independent mechanism. Astrocyte-oligodendrocyte cocultures exposed to AQP4-IgG and complement showed prominent MAC deposition on oligodendrocytes in contact with astrocytes, whereas C1q, the initiating protein in the classical complement pathway, and C3d, a component of the alternative complement pathway, were deposited only on astrocytes. Early oligodendrocyte injury with MAC deposition was also found in rat brain following intracerebral injection of AQP4-IgG, complement and a fixable dead-cell stain. These results support a novel complement bystander mechanism for early oligodendrocyte injury and demyelination in NMO.


Assuntos
Aquaporina 4/imunologia , Astrócitos/imunologia , Efeito Espectador/imunologia , Proteínas do Sistema Complemento/metabolismo , Neuromielite Óptica/imunologia , Oligodendroglia/imunologia , Animais , Aquaporina 4/genética , Astrócitos/patologia , Autoanticorpos/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Imunoglobulina G/imunologia , Neuromielite Óptica/patologia , Oligodendroglia/patologia , Ratos Sprague-Dawley , Ratos Transgênicos , Proteínas Recombinantes/imunologia
4.
Adv Exp Med Biol ; 969: 239-250, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28258578

RESUMO

Drugs targeting aquaporins have broad potential clinical applications, including cancer, obesity, edema, glaucoma, skin diseases and others. The astrocyte water channel aquaporin-4 is a particularly compelling target because of its role of brain water movement, neuroexcitation and glia scarring, and because it is the target of pathogenic autoantibodies in the neuroinflammatory demyelinating disease neuromyelitis optica . There has been considerable interest in the identification of small molecule inhibitors of aquaporins, with various candidates emerging from testing of known ion transport inhibitors, as well as compound screening and computational chemistry. However, in general, the activity of reported aquaporin inhibitors has not been confirmed on retesting, which may be due to technical problems in water transport assays used in the original identification studies, and the challenges in modulating the activity of small, compact, pore-containing membrane proteins. We review here the state of the field of aquaporin-modulating small molecules and biologics, and the challenges and opportunities in moving forward.


Assuntos
Aquaporina 4/antagonistas & inibidores , Edema Encefálico/tratamento farmacológico , Neuromielite Óptica/tratamento farmacológico , Tiadiazóis/farmacologia , Água/metabolismo , Animais , Aquaporina 4/genética , Aquaporina 4/metabolismo , Transporte Biológico , Edema Encefálico/genética , Edema Encefálico/metabolismo , Edema Encefálico/patologia , Regulação da Expressão Gênica , Humanos , Terapia de Alvo Molecular , Neuromielite Óptica/genética , Neuromielite Óptica/metabolismo , Neuromielite Óptica/patologia , Concentração Osmolar , Pressão Osmótica , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sumatriptana/farmacologia , Triazóis/farmacologia , Triptaminas/farmacologia
5.
Pediatr Neurosurg ; 52(6): 409-416, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27978530

RESUMO

The aquaporins (AQPs) are a family of water-transporting proteins that are broadly expressed in mammalian cells. Two AQPs in the central nervous system, AQP1 and AQP4, might play a role in hydrocephalus and are thus potential drug targets. AQP1 is expressed in the ventricular-facing membrane of choroid plexus epithelial cells, where it facilitates the secretion of cerebrospinal fluid (CSF). AQP4 is expressed in astrocyte foot processes and ependymal cells lining ventricles, where it appears to facilitate the transport of excess water out of the brain. Altered expression of these AQPs in experimental animal models of hydrocephalus and limited human specimens suggests their involvement in the pathophysiology of hydrocephalus, as do data in knockout mice demonstrating a protective effect of AQP1 deletion and a deleterious effect of AQP4 deletion in hydrocephalus. Though significant questions remain, including the precise contribution of AQP1 to CSF secretion in humans and the mechanisms by which AQP4 facilitates clearance of excess brain water, AQP1 and AQP4 have been proposed as potential drug targets to reduce ventricular enlargement in hydrocephalus.


Assuntos
Aquaporinas/metabolismo , Hidrocefalia/metabolismo , Animais , Aquaporinas/fisiologia , Água Corporal/fisiologia , Encéfalo/metabolismo , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Plexo Corióideo/metabolismo , Humanos
6.
Clin Gastroenterol Hepatol ; 12(2): 204-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24316107

RESUMO

Diarrheal diseases constitute a significant global health burden and are a major cause of childhood mortality and morbidity. Treatment of diarrheal disease has centered on the replacement of fluid and electrolyte losses using oral rehydration solutions. Although oral rehydration solutions have been highly successful, significant mortality and morbidity due to diarrheal disease remains. Secretory diarrheas, such as those caused by bacterial and viral enterotoxins, result from activation of cyclic nucleotide and/or Ca(2+) signaling pathways in intestinal epithelial cells, enterocytes, which increase the permeability of Cl(-) channels at the lumen-facing membrane. Additionally, there is often a parallel reduction in intestinal Na(+) absorption. Inhibition of enterocyte Cl(-) channels, including the cystic fibrosis transmembrane conductance regulator and Ca(2+)-activated Cl(-) channels, represents an attractive strategy for antisecretory drug therapy. High-throughput screening of synthetic small-molecule collections has identified several classes of Cl(-) channel inhibitors that show efficacy in animal models of diarrhea but remain to be tested clinically. In addition, several natural product extracts with Cl(-) channel inhibition activity have shown efficacy in diarrhea models. However, a number of challenges remain to translate the promising bench science into clinically useful therapeutics, including efficiently targeting orally administered drugs to enterocytes during diarrhea, funding development costs, and carrying out informative clinical trials. Nonetheless, Cl(-) channel inhibitors may prove to be effective adjunctive therapy in a broad spectrum of clinical diarrheas, including acute infectious and drug-related diarrheas, short bowel syndrome, and congenital enteropathies.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Diarreia/tratamento farmacológico , Animais , Antidiarreicos/farmacologia , Transporte Biológico/efeitos dos fármacos , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Diarreia/metabolismo , Diarreia/microbiologia , Modelos Animais de Doenças , Humanos
7.
Ann Neurol ; 73(1): 77-85, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23055279

RESUMO

OBJECTIVE: Neuromyelitis optica (NMO) is caused by binding of pathogenic autoantibodies (NMO-immunoglobulin G [IgG]) to aquaporin-4 (AQP4) on astrocytes, which initiates complement-dependent cytotoxicity (CDC) and inflammation. We recently introduced mutated antibody (aquaporumab) and small-molecule blocker strategies for therapy of NMO, based on prevention of NMO-IgG binding to AQP4. Here, we investigated an alternative strategy involving neutralization of NMO-IgG effector function by selective IgG heavy-chain deglycosylation with bacteria-derived endoglycosidase S (EndoS). METHODS: Cytotoxicity and NMO pathology were measured in cell and spinal cord slice cultures, and in mice exposed to control or EndoS-treated NMO-IgG. RESULTS: EndoS treatment of NMO patient serum reduced by >95% CDC and antibody-dependent cell-mediated cytotoxicity, without impairment of NMO-IgG binding to AQP4. Cytotoxicity was also prevented by addition of EndoS after NMO-IgG binding to AQP4. The EndoS-treated, nonpathogenic NMO-IgG competitively displaced pathogenic NMO-IgG bound to AQP4, and prevented NMO pathology in spinal cord slice culture and mouse models of NMO. INTERPRETATION: EndoS deglycosylation converts pathogenic NMO-IgG autoantibodies into therapeutic blocking antibodies. EndoS treatment of blood may be beneficial in NMO, and may be accomplished, for example, by therapeutic apheresis using surface-immobilized EndoS.


Assuntos
Aquaporina 4/sangue , Aquaporina 4/uso terapêutico , Imunoglobulina G/biossíntese , Neuromielite Óptica/sangue , Neuromielite Óptica/terapia , Animais , Autoanticorpos/biossíntese , Autoanticorpos/sangue , Autoanticorpos/uso terapêutico , Proteínas de Bactérias/fisiologia , Células CHO , Cricetinae , Cricetulus , Glicosídeo Hidrolases/fisiologia , Glicosilação , Humanos , Imunoglobulina G/sangue , Camundongos , Camundongos Knockout , Técnicas de Cultura de Órgãos , Medula Espinal/enzimologia
8.
Mol Pharmacol ; 83(6): 1268-75, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23571414

RESUMO

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.


Assuntos
Aquaporina 4/imunologia , Autoanticorpos/metabolismo , Proteínas de Bactérias/farmacologia , Cisteína Endopeptidases/farmacologia , Citotoxicidade Imunológica , Imunoglobulina G/metabolismo , Neuromielite Óptica/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos , Proteínas de Bactérias/uso terapêutico , Células CHO , Proteínas do Sistema Complemento/imunologia , Cricetinae , Cricetulus , Cisteína Endopeptidases/uso terapêutico , Humanos , Imunoglobulina G/imunologia , Camundongos , Neuromielite Óptica/patologia , Neuromielite Óptica/terapia
9.
J Biol Chem ; 287(17): 13829-39, 2012 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-22393049

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system in which binding of pathogenic autoantibodies (NMO-IgG) to astrocyte aquaporin-4 (AQP4) causes complement-dependent cytotoxicity (CDC) and inflammation. We previously reported a wide range of binding affinities of NMO-IgGs to AQP4 in separate tetramers versus intramembrane aggregates (orthogonal arrays of particles, OAPs). We report here a second, independent mechanism by which CDC is affected by AQP4 assembly. Utilizing lactate dehydrogenase release and live/dead cell cytotoxicity assays, we found in different cell lines, and with different monoclonal and patient-derived NMO-IgGs, that CDC was greatly (>100-fold) reduced in cells expressing M1- versus M23-AQP4. Studies using a M23-AQP4 mutant containing an OAP-disrupting mutation, and in cells expressing AQP4 in different M1/M23 ratios, indicated that NMO-IgG-dependent CDC requires AQP4 OAP assembly. In contrast, antibody-dependent cell-mediated cytotoxicity produced by natural killer cells did not depend on AQP4 OAP assembly. Measurements of C1q binding and complement attack complex (C9neo) supported the conclusion that the greatly enhanced CDC by OAPs is due to efficient, multivalent binding of C1q to clustered NMO-IgG on OAPs. We conclude that AQP4 assembly in OAPs is required for CDC in NMO, establishing a new mechanism of OAP-dependent NMO pathogenesis. Disruption of AQP4 OAPs may greatly reduce NMO-IgG dependent CDC and NMO pathology.


Assuntos
Aquaporina 4/metabolismo , Neuromielite Óptica/metabolismo , Animais , Astrócitos/citologia , Autoanticorpos/química , Células CHO , Linhagem Celular Tumoral , Ativação do Complemento , Complemento C1q/química , Proteínas do Sistema Complemento , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/química , Ligação Proteica
10.
J Biol Chem ; 287(44): 36837-44, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22989877

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system caused by binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to AQP4 on astrocytes. A screen was developed to identify inhibitors of NMO-IgG-dependent, complement-dependent cytotoxicity. Screening of 50,000 synthetic small molecules was done using CHO cells expressing human AQP4 and a human NMO recombinant monoclonal antibody (rAb-53). The screen yielded pyrano[2,3-c]pyrazoles that blocked rAb-53 binding to AQP4 and prevented cytotoxicity in cell culture and spinal cord slice models of NMO. Structure-activity analysis of 82 analogs yielded a blocker with IC(50) ∼ 6 µm. Analysis of the blocker mechanism indicated idiotype specificity, as (i) pyrano[2,3-c]pyrazoles did not prevent AQP4 binding or cytotoxicity of other NMO-IgGs, and (ii) surface plasmon resonance showed specific rAb-53 binding. Antibody structure modeling and docking suggested a putative binding site near the complementarity-determining regions. Small molecules with idiotype-specific antibody targeting may be useful as research tools and therapeutics.


Assuntos
Aquaporina 4/imunologia , Autoanticorpos/metabolismo , Imunoglobulina G/metabolismo , Neuromielite Óptica/imunologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Aquaporina 4/metabolismo , Autoanticorpos/imunologia , Sítios de Ligação , Células CHO , Cricetinae , Ensaios de Triagem em Larga Escala , Humanos , Imunoglobulina G/imunologia , Idiótipos de Imunoglobulinas/imunologia , Idiótipos de Imunoglobulinas/metabolismo , Camundongos , Camundongos Knockout , Simulação de Acoplamento Molecular , Neuromielite Óptica/metabolismo , Ligação Proteica/efeitos dos fármacos , Piranos/farmacologia , Pirazóis/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície , Técnicas de Cultura de Tecidos
11.
Ann Neurol ; 71(3): 314-22, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22271321

RESUMO

OBJECTIVE: Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of the central nervous system. Circulating autoantibodies (NMO-immunoglobulin [Ig]G) against astrocyte water channel aquaporin-4 (AQP4) cause complement- and cell-mediated astrocyte damage with consequent neuroinflammation and demyelination. Current NMO therapies, which have limited efficacy, include immunosuppression and plasma exchange. The objective of this study was to develop a potential new NMO therapy based on blocking of pathogenic NMO-IgG binding to its target, AQP4. METHODS: We generated nonpathogenic recombinant monoclonal anti-AQP4 antibodies that selectively block NMO-IgG binding to AQP4. These antibodies comprise a tight-binding anti-AQP4 Fab and a mutated Fc that lacks functionality for complement- and cell-mediated cytotoxicity. The efficacy of the blocking antibodies was studied using cell culture, spinal cord slice, and in vivo mouse models of NMO. RESULTS: In AQP4-expressing cell cultures, the nonpathogenic competing antibodies blocked binding of NMO-IgG in human sera, reducing to near zero complement- and cell-mediated cytotoxicity. The antibodies prevented the development of NMO lesions in an ex vivo spinal cord slice model of NMO and in an in vivo mouse model, without causing cytotoxicity. INTERPRETATION: Our results provide proof of concept for a therapy of NMO with blocking antibodies. The broad efficacy of antibody inhibition is likely due to steric competition because of its large physical size compared to AQP4. Blocker therapy to prevent binding of pathogenic autoantibodies to their targets may be useful for treatment of other autoimmune diseases as well.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Aquaporina 4/antagonistas & inibidores , Aquaporina 4/metabolismo , Neuromielite Óptica/tratamento farmacológico , Neuromielite Óptica/metabolismo , Animais , Animais Recém-Nascidos , Aquaporina 4/imunologia , Sítios de Ligação de Anticorpos , Ligação Competitiva/imunologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/metabolismo , Camundongos , Camundongos Knockout , Neuromielite Óptica/imunologia , Técnicas de Cultura de Órgãos , Ligação Proteica/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico
12.
Acta Neuropathol ; 125(6): 829-40, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23677375

RESUMO

Neuromyelitis optica (NMO) is an autoimmune disorder with inflammatory demyelinating lesions in the central nervous system, particularly in the spinal cord and optic nerve. NMO pathogenesis is thought to involve binding of anti-aquaporin-4 (AQP4) autoantibodies to astrocytes, which causes complement-dependent cytotoxicity (CDC) and downstream inflammation leading to oligodendrocyte and neuronal injury. Vasculocentric deposition of activated complement is a prominent feature of NMO pathology. Here, we show that a neutralizing monoclonal antibody against the C1q protein in the classical complement pathway prevents AQP4 autoantibody-dependent CDC in cell cultures and NMO lesions in ex vivo spinal cord slice cultures and in mice. A monoclonal antibody against human C1q with 11 nM binding affinity prevented CDC caused by NMO patient serum in AQP4-transfected cells and primary astrocyte cultures, and prevented complement-dependent cell-mediated cytotoxicity (CDCC) produced by natural killer cells. The anti-C1q antibody prevented astrocyte damage and demyelination in mouse spinal cord slice cultures exposed to AQP4 autoantibody and human complement. In a mouse model of NMO produced by intracerebral injection of AQP4 autoantibody and human complement, the inflammatory demyelinating lesions were greatly reduced by intracerebral administration of the anti-C1q antibody. These results provide proof-of-concept for C1q-targeted monoclonal antibody therapy in NMO. Targeting of C1q inhibits the classical complement pathway directly and causes secondary inhibition of CDCC and the alternative complement pathway. As C1q-targeted therapy leaves the lectin complement activation pathway largely intact, its side-effect profile is predicted to differ from that of therapies targeting downstream complement proteins.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complemento C1q/antagonistas & inibidores , Fatores Imunológicos/uso terapêutico , Neuromielite Óptica/patologia , Neuromielite Óptica/prevenção & controle , Animais , Aquaporina 4/fisiologia , Técnicas de Cultura de Células , Ativação do Complemento , Cricetulus , Modelos Animais de Doenças , Humanos , Camundongos , Neuromielite Óptica/etiologia
13.
FASEB J ; 26(5): 2197-208, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22319008

RESUMO

Neuromyelitis optica (NMO) is an inflammatory demyelinating disease of spinal cord and optic nerve caused by pathogenic autoantibodies (NMO-IgG) against astrocyte aquaporin-4 (AQP4). We developed a high-throughput screen to identify blockers of NMO-IgG binding to human AQP4 using a human recombinant monoclonal NMO-IgG and transfected Fisher rat thyroid cells stably expressing human M23-AQP4. Screening of ∼60,000 compounds yielded the antiviral arbidol, the flavonoid tamarixetin, and several plant-derived berbamine alkaloids, each of which blocked NMO-IgG binding to AQP4 without affecting AQP4 expression, array assembly, or water permeability. The compounds inhibited NMO-IgG binding to AQP4 in NMO patient sera and blocked NMO-IgG-dependent complement- and cell-mediated cytotoxicity with IC(50) down to ∼5 µM. Docking computations identified putative sites of blocker binding at the extracellular surface of AQP4. The blockers did not affect complement-dependent cytotoxicity caused by anti-GD3 antibody binding to ganglioside GD3. The blockers reduced by >80% the severity of NMO lesions in an ex vivo spinal cord slice culture model of NMO and in mice in vivo. Our results provide proof of concept for a small-molecule blocker strategy to reduce NMO pathology. Small-molecule blockers may also be useful for other autoimmune diseases caused by binding of pathogenic autoantibodies to defined targets.


Assuntos
Aquaporina 4/metabolismo , Astrócitos/metabolismo , Imunoglobulina G/metabolismo , Neuromielite Óptica/patologia , Bibliotecas de Moléculas Pequenas , Animais , Camundongos , Ratos , Ratos Endogâmicos F344
15.
Acta Pharmacol Sin ; 32(6): 702-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21552296

RESUMO

Aquaporin-4 (AQP4) is a water-selective transporter expressed in astrocytes throughout the central nervous system, as well as in kidney, lung, stomach and skeletal muscle. The two AQP4 isoforms produced by alternative spicing, M1 and M23 AQP4, form heterotetramers that assemble in cell plasma membranes in supramolecular structures called orthogonal arrays of particles (OAPs). Phenotype analysis of AQP4-null mice indicates the involvement of AQP4 in brain and spinal cord water balance, astrocyte migration, neural signal transduction and neuroinflammation. AQP4-null mice manifest reduced brain swelling in cytotoxic cerebral edema, but increased brain swelling in vasogenic edema and hydrocephalus. AQP4 deficiency also increases seizure duration, impairs glial scarring, and reduces the severity of autoimmune neuroinflammation. Each of these phenotypes is likely explicable on the basis of reduced astrocyte water permeability in AQP4 deficiency. AQP4 is also involved in the neuroinflammatory demyelinating disease neuromyelitis optica (NMO), where autoantibodies (NMO-IgG) targeting AQP4 produce astrocyte damage and inflammation. Mice administered NMO-IgG and human complement by intracerebral injection develop characteristic NMO lesions with neuroinflammation, demyelination, perivascular complement deposition and loss of glial fibrillary acidic protein and AQP4 immunoreactivity. Our findings suggest the potential utility of AQP4-based therapeutics, including small-molecule modulators of AQP4 water transport function for therapy of brain swelling, injury and epilepsy, as well as small-molecule or monoclonal antibody blockers of NMO-IgG binding to AQP4 for therapy of NMO.


Assuntos
Aquaporina 4 , Sistema Nervoso Central/fisiologia , Descoberta de Drogas , Neuromielite Óptica/tratamento farmacológico , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Aquaporina 4/química , Aquaporina 4/imunologia , Aquaporina 4/fisiologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Dados de Sequência Molecular , Neuromielite Óptica/imunologia , Neuromielite Óptica/metabolismo , Ligação Proteica , Multimerização Proteica , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/uso terapêutico
16.
Sci Rep ; 11(1): 21962, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34753987

RESUMO

Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system. Most NMOSD patients are seropositive for immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4), called AQP4-IgG. AQP4-IgG binding to aquaporin-4 causes complement-dependent cytotoxicity (CDC), leading to inflammation and demyelination. Here, CDC was measured in AQP4-expressing cells exposed to human complement and heat-inactivated sera from 108 AQP4-IgG seropositive NMOSD subjects and 25 non-NMOSD controls. AQP4-IgG positive sera produced a wide range of CDC, with 50% maximum cytotoxicity produced by as low as 0.2% serum concentration. Unexpectedly, 58 samples produced no cytotoxicity, and of those, four sera were cytoprotective against cytotoxic AQP4-IgG. Cytoprotection was found against different cytotoxic monoclonal AQP4-IgGs and NMOSD patient sera, and in primary astrocyte cultures. Mechanistic studies revealed that the protective factor is an IgG antibody that did not inhibit complement directly, but interfered with binding of cytotoxic AQP4-IgG to AQP4 and consequent C1q binding and complement activation. Further studies suggested that non-pathogenic AQP4-IgG, perhaps with altered glycosylation, may contribute to reduced or ineffectual binding of cytotoxic AQP4-IgG, as well as reduced cell-surface AQP4. The presence of natural cytoprotective antibodies in AQP4-IgG seropositive sera reveals an added level of complexity in NMOSD disease pathogenesis, and suggests the potential therapeutic utility of 'convalescent' serum or engineered protective antibody to interfere with pathogenic antibody in AQP4-IgG seropositive NMOSD.


Assuntos
Aquaporina 4/imunologia , Neuromielite Óptica/imunologia , Animais , Aquaporina 4/sangue , Autoanticorpos/sangue , Autoanticorpos/imunologia , Biomarcadores/sangue , Células CHO , Cricetulus , Progressão da Doença , Humanos , Soros Imunes , Imunoglobulina G/sangue , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia
17.
Expert Opin Biol Ther ; 21(8): 1073-1086, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33513036

RESUMO

INTRODUCTION: Neuromyelitis optica spectrum disorder (NMOSD) is characterized by central nervous system inflammation and demyelination. In AQP4-IgG seropositive NMOSD, circulating immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4) cause tissue injury. Compelling evidence supports a pathogenic role for complement activation following AQP4-IgG binding to AQP4. Clinical studies supported the approval of eculizumab, an inhibitor of C5 cleavage, in AQP4-IgG seropositive NMOSD. AREAS COVERED: This review covers in vitro, animal models, and human evidence for complement-dependent and complement-independent tissue injury in AQP4-IgG seropositive NMOSD. Complement targets are discussed, including complement proteins, regulators and anaphylatoxin receptors, and corresponding drug candidates. EXPERT OPINION: Though preclinical data support a central pathogenic role of complement activation in AQP4-IgG seropositive NMOSD, they do not resolve the relative contributions of complement-dependent vs. complement-independent disease mechanisms such as antibody-dependent cellular cytotoxicity, T cell effector mechanisms, and direct AQP4-IgG-induced cellular injury. The best evidence that complement-dependent mechanisms predominate in AQP4-IgG seropositive NMOSD comes from eculizumab clinical data. Various drug candidates targeting distinct complement effector mechanisms may offer improved safety and efficacy. However, notwithstanding the demonstrated efficacy of complement inhibition in AQP4-IgG seropositive NMOSD, the ultimate niche for complement inhibition is not clear given multiple drug options with alternative mechanisms of action.Abbreviations: AAV2, Adeno-associated virus 2; ADCC, antibody-dependent cellular cytotoxicity; ANCA, antineutrophilic cytoplasmic autoantibody; AQP4, aquaporin-4; AQP4-IgG, AQP4-immunoglobulin G; C1-INH, C1-esterase inhibitor; C3aR, C3a receptor; C4BP, C4 binding protein; C5aR, C5a receptor; CDC, complement-dependent cytotoxicity; CFHR1, complement factor H related 1; CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; EndoS, endoglycosidase S; FHL-1, factor-H-like protein 1; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor protein-1; IgG, immunoglobulin G; IVIG, intravenous human immunoglobulin G; MAC, membrane attack complex; MBL, maltose-binding lectin; MBP, myelin basic protein; MOG, myelin oligodendrocyte glycoprotein; NK cell, natural killer cell; NMOSD, neuromyelitis optica spectrum disorder; OAP, orthogonal arrays of particles; PNH, paroxysmal nocturnal hemoglobinuria.


Assuntos
Neuromielite Óptica , Animais , Aquaporina 4 , Autoanticorpos , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunoglobulina G , Neuromielite Óptica/tratamento farmacológico
18.
Mol Pharmacol ; 77(1): 69-78, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19808995

RESUMO

Crofelemer, a purified proanthocyanidin oligomer extracted from the bark latex of Croton lechleri, is in clinical trials for secretory diarrheas of various etiologies. We investigated the antisecretory mechanism of crofelemer by determining its effect on the major apical membrane transport and signaling processes involved in intestinal fluid transport. Using cell lines and measurement procedures to isolate the effects on individual membrane transport proteins, crofelemer at 50 microM had little or no effect on the activity of epithelial Na(+) or K(+) channels or on cAMP or calcium signaling. Crofelemer inhibited the cystic fibrosis transmembrane regulator (CFTR) Cl(-) channel with maximum inhibition of approximately 60% and an IC(50) approximately 7 microM. Crofelemer action at an extracellular site on CFTR produced voltage-independent block with stabilization of the channel closed state. Crofelemer did not affect the potency of glycine hydrazide or thiazolidinone CFTR inhibitors. Crofelemer action resisted washout, with <50% reversal of CFTR inhibition after 4 h. Crofelemer was also found to strongly inhibit the intestinal calcium-activated Cl(-) channel TMEM16A by a voltage-independent inhibition mechanism with maximum inhibition >90% and IC(50) approximately 6.5 microM. The dual inhibitory action of crofelemer on two structurally unrelated prosecretory intestinal Cl(-) channels may account for its intestinal antisecretory activity.


Assuntos
Antidiarreicos/farmacologia , Canais de Cloreto/antagonistas & inibidores , Croton/química , Mucosa Intestinal/metabolismo , Proantocianidinas/farmacologia , Anoctamina-1 , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Humanos , Concentração Inibidora 50 , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Proantocianidinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
19.
Expert Opin Ther Targets ; 24(3): 219-229, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32070155

RESUMO

Introduction: Neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelinating disease of the central nervous system affecting primarily the spinal cord and optic nerves. Most NMOSD patients are seropositive for immunoglobulin G autoantibodies against astrocyte water channel aquaporin-4, called AQP4-IgG, which cause astrocyte injury leading to demyelination and neurological impairment. Current therapy for AQP4-IgG seropositive NMOSD includes immunosuppression, B cell depletion, and plasma exchange. Newer therapies target complement, CD19 and IL-6 receptors.Areas covered: This review covers early-stage pre-clinical therapeutic approaches for seropositive NMOSD. Targets include pathogenic AQP4-IgG autoantibodies and their binding to AQP4, complement-dependent and cell-mediated cytotoxicity, blood-brain barrier, remyelination and immune effector and regulatory cells, with treatment modalities including small molecules, biologics, and cells.Expert opinion: Though newer NMOSD therapies appear to have increased efficacy in reducing relapse rate and neurological deficit, increasingly targeted therapies could benefit NMOSD patients with ongoing relapses and could potentially be superior in efficacy and safety. Of the various early-stage therapeutic approaches, IgG inactivating enzymes, aquaporumab blocking antibodies, drugs targeting early components of the classical complement system, complement regulator-targeted drugs, and Fc-based multimers are of interest. Curative strategies, perhaps involving AQP4 tolerization, remain intriguing future possibilities.


Assuntos
Desenvolvimento de Medicamentos , Terapia de Alvo Molecular , Neuromielite Óptica/tratamento farmacológico , Animais , Aquaporina 4/imunologia , Autoanticorpos/imunologia , Proteínas do Sistema Complemento/metabolismo , Humanos , Imunoglobulina G/imunologia , Neuromielite Óptica/imunologia , Neuromielite Óptica/fisiopatologia
20.
Neuropharmacology ; 162: 107827, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31654702

RESUMO

Pathogenesis in seropositive neuromyelitis optica spectrum disorders (herein called NMO) involves binding of IgG1 autoantibodies to aquaporin-4 (AQP4) on astrocytes in the central nervous system, which initiates complement and cellular injury. We previously developed an antibody blocking approach for potential therapy of NMO in which an engineered, monoclonal, anti-AQP4 antibody lacking cytotoxicity effector functions (called aquaporumab) blocked binding of NMO autoantibodies to astrocyte AQP4 (Tradtrantip et al. Ann. Neurol. 71, 314-322, 2012). Here, a high-affinity aquaporumab, which was generated by affinity maturation using saturation mutagenesis, was shown to block cellular injury caused by NMO patient sera. Anti-AQP4 antibody rAb-53, a fully human antibody with effector function neutralizing Fc mutations L234A/L235A and affinity-enhancing Fab mutations Y50R/S56R, called AQmabAM, bound to AQP4 in cell cultures with Kd ~ 18 ng/ml (~0.12 nM), ~8-fold greater affinity than the original antibody. AQmabAM, but without L234A/L235A Fc mutations, produced complement-dependent cytotoxicity (CDC) with EC50 ~ 82 ng/ml. AQmabAM prevented CDC produced by sera from eight NMO patients with IC50 ranging from 40 to 80 ng/ml, and similarly prevented antibody-dependent cellular cytotoxicity (ADCC). Mechanistic studies demonstrated that AQmabAM blocked binding of serum NMO autoantibodies to AQP4. AQmabAM offers a targeted, non-immunosuppressive approach for therapy of seropositive NMO. Autoantibody blocking may be a useful therapeutic strategy for other autoimmune diseases as well.


Assuntos
Anticorpos Bloqueadores/farmacologia , Afinidade de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Aquaporina 4/imunologia , Autoanticorpos/imunologia , Imunoglobulina G/farmacologia , Neuromielite Óptica/tratamento farmacológico , Proteínas Recombinantes/farmacologia , Animais , Anticorpos Bloqueadores/uso terapêutico , Anticorpos Monoclonais , Afinidade de Anticorpos/genética , Citotoxicidade Celular Dependente de Anticorpos/genética , Ligação Competitiva , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Cricetulus , Testes Imunológicos de Citotoxicidade , Humanos , Imunoglobulina G/genética , Células Matadoras Naturais , Mutagênese , Neuromielite Óptica/imunologia , Soro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA