Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncoimmunology ; 9(1): 1744980, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32363112

RESUMO

ADAM10 and ADAM17 expression and soluble PD-L1 (sPD-L1) predict poor prognosis in many malignancies, including in patients treated with PD-(L)1 inhibitors. The mechanism of soluble PD-L1 production and its effects are unknown. Here we uncover a novel mechanism of ADAM10- and ADAM17-mediated resistance to PD-(L)1 inhibitors. ADAM10 and ADAM17 cleave PD-L1 from the surface of malignant cells and extracellular vesicles. This cleavage produces an active sPD-L1 fragment that induces apoptosis in CD8 + T cells and compromises the killing of tumor cells by CD8 + T cells. Reduced tumor site PD-L1 protein-to-mRNA ratios predict poor outcomes and are correlated with elevated ADAM10 and ADAM17 expression in multiple cancers. These results may explain the discordance between PD-L1 immunohistochemistry and PD-(L)1 inhibitor response. Thus, including ADAM10 and ADAM17 tissue staining may improve therapy selection. Furthermore, treatment with an ADAM10/ADAM17 inhibitor may abrogate PD-(L)1 inhibitor resistance and improve clinical responses to PD-(L)1 immunotherapy.


Assuntos
Secretases da Proteína Precursora do Amiloide , Antígeno B7-H1 , Proteína ADAM10 , Proteína ADAM17 , Apoptose , Antígeno B7-H1/genética , Humanos , Proteínas de Membrana/genética
2.
Cell Immunol ; 347: 104020, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31767118

RESUMO

High-mobility group box 1 (HMGB1) concentration in serum or plasma has been proposed as an important biological marker in various inflammation-related pathologies. We previously showed that low titer autoantibodies against HMGB1 could emerge during the course of sepsis. Importantly their presence was positively related with patients' survival. In this study, we focused on plasma samples from 2 patients who survived sepsis and exhibited high titer antibodies to HMGB1. These antibodies were proved to be specific for HMGB1 since they did not bind to HMGB2 or to human serum albumin. Following IgG purification, it has shown that both patients secreted HMGB1-hydrolyzing autoantibodies in vitro. These findings suggested that proteolytic antibodies directed against HMGB1 can be produced in patients surviving septic shock.


Assuntos
Autoanticorpos/imunologia , Proteína HMGB1/imunologia , Choque Séptico/imunologia , Autoanticorpos/sangue , Proteína HMGB2/imunologia , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Proteólise , Albumina Sérica Humana/imunologia , Choque Séptico/mortalidade , Choque Séptico/patologia
3.
Expert Opin Biol Ther ; 18(5): 545-560, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29460650

RESUMO

INTRODUCTION: Multiple sclerosis (MS) is a chronic and progressive inflammatory demyelinating disease of the human central nervous system (CNS) and is the most common disabling neurological condition in young adults, resulting in severe neurological defects. No curative or long-term progression-inhibiting therapy has yet been developed. However, recent investigation has revealed potential strategies that do not merely modulate potentially pathogenic autoimmune responses, but stimulate remyelination within CNS lesions. AREAS COVERED: We discuss the history and development of natural human IgM-isotype immunoglobulins (HIgMs) and recently-identified aptamer-conjugates that have been shown to enhance endogenous myelin repair in animal models of demyelination by acting on myelin-producing oligodendrocytes (OLs) or oligodendrocyte progenitor cells (OPCs) within CNS lesions. We also discuss future development aims and applications for these important novel technologies. EXPERT OPINION: Aptamer conjugate Myaptavin-3064 and recombinant human IgM-isotype antibody rHIgM22 regenerate CNS myelin, thereby reducing axonal degeneration and offering the potential of recovery from MS relapses, reversal of disability and prevention of disease progression. Advancement of these technologies into the clinic for MS treatment is therefore a top priority. It remains unclear to what extent the therapeutic modalities of remyelinating antibodies and aptamers may synergize with other currently-approved therapies to yield enhanced therapeutic effects.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Produtos Biológicos/uso terapêutico , Doenças do Sistema Nervoso Central/tratamento farmacológico , Imunoconjugados/uso terapêutico , Remielinização/efeitos dos fármacos , Adulto , Animais , Aptâmeros de Peptídeos/uso terapêutico , Doenças Desmielinizantes/tratamento farmacológico , Humanos , Esclerose Múltipla/tratamento farmacológico , Regeneração/efeitos dos fármacos , Remielinização/fisiologia , Adulto Jovem
4.
Hum Antibodies ; 25(3-4): 121-129, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28269761

RESUMO

A single peripheral dose of CNS-binding IgMs promote remyelination and preserve axons in a number of animal models of neurologic disease. A myelin-binding recombinant human IgM (rHIgM22) is presently in a safety trial in MS patients following an acute MS exacerbation. rHIgM22 (directed against oligodendrocytes) or rHIgM12 (directed against neurons) were administered to mice with MOG-induced experimental autoimmune encephalomyelitis (EAE) with study endpoints: clinical deficits and brain and spinal cord pathology. IgMs were administered at a therapeutic dose of 100 µ g intra peritoneal at the time of immunization (day -1, 0, +$1), disease onset (15 days) or peak of the disease (28 days). Disease course was not worsened by either human IgM regardless of the time of treatment. Of note, the human IgM that recognizes a carbohydrate epitope on gangliosides and NCAM, rHIgM12, reduced brain pathology when given at time of immunization or at onset of disease, but did not reduce clinical deficits or spinal cord disease burden. Hence, treatment with rHIgM12 resulted in marked reduction in meningeal inflammation. Data consistent with the hypothesis that in the EAE model this molecule has an immune-modulatory effect. Treatment with an anti-CD4 blocking IgG prevented both clinical course and CNS pathology. This pre-clinical study further supports the safety of therapeutic CNS-binding human IgMs in the presence of autoimmunity and clearly differentiates them from IgGs directed against MOG or aquaporin-4 that worsen neurologic disease.


Assuntos
Disfunção Cognitiva/tratamento farmacológico , Doenças Desmielinizantes/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Imunoglobulina M/farmacologia , Fatores Imunológicos/farmacologia , Molécula L1 de Adesão de Célula Nervosa/imunologia , Fármacos Neuroprotetores/farmacologia , Ácidos Siálicos/imunologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/patologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Esquema de Medicação , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Adjuvante de Freund/administração & dosagem , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Glicoproteína Mielina-Oligodendrócito/administração & dosagem , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/imunologia , Oligodendroglia/patologia , Fragmentos de Peptídeos/administração & dosagem , Ligação Proteica , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/metabolismo , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia
5.
J Neuroinflammation ; 13(1): 293, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27855706

RESUMO

BACKGROUND: We investigated the role of human HLA class I molecules in persistent central nervous system (CNS) injury versus repair following virus infection of the CNS. METHODS: Human class I A11+ and B27+ transgenic human beta-2 microglobulin positive (Hß2m+) mice of the H-2 b background were generated on a combined class I-deficient (mouse beta-2 microglobulin deficient, ß2m0) and class II-deficient (mouse Aß0) phenotype. Intracranial infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL mice results in acute encephalitis with prominent injury in the hippocampus, striatum, and cortex. RESULTS: Following infection with TMEV, a picornavirus, the Aß0.ß2m0 mice lacking active immune responses died within 18 to 21 days post-infection. These mice showed severe encephalomyelitis due to rapid replication of the viral genome. In contrast, transgenic Hß2m mice with insertion of a single human class I MHC gene in the absence of human or mouse class II survived the acute infection. Both A11+ and B27+ mice significantly controlled virus RNA expression by 45 days and did not develop late-onset spinal cord demyelination. By 45 days post-infection (DPI), B27+ transgenic mice showed almost complete repair of the virus-induced brain injury, but A11+ mice conversely showed persistent severe hippocampal and cortical injury. CONCLUSIONS: The findings support the hypothesis that the expression of a single human class I MHC molecule, independent of persistent virus infection, influences the extent of sub frequent chronic neuronal injury or repair in the absence of a class II MHC immune response.


Assuntos
Infecções por Cardiovirus/patologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Theilovirus/fisiologia , Análise de Variância , Animais , Anticorpos/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Antígeno HLA-A11/metabolismo , Antígeno HLA-B27/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
6.
Int J Phys Med Rehabil ; 4(4)2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27595123

RESUMO

We provide an overview of rehabilitation in neurological diseases. A large amount of literature available on neurorehabilitation is based from the rehabilitative work on stroke and spinal cord injuries. After a brief description of rehabilitation, the potential application of neurorehabilitation in neurodegenerative diseases specifically multiple sclerosis (MS) is summarized. Since MS causes a wide variety of symptoms, the rehabilitation in MS patients may benefit from an interdisciplinary approach that encloses physiotherapy, cognitive rehabilitation, psychological therapy, occupational therapy, and other methods to improve fatigue. Neurorehabilitation helps patients to reach and maintain their optimal physical, psychological and intellectual, levels but it does not reverse long-term disabilities that arise from neurological disorders. This calls for the need of better neuroregenerative and neuroprotective treatment strategies in addition to neurorehabilitation. We discuss neuroprotective drugs aimed at preventing axonal, neuronal, myelin and oligodendrocyte damage and cell death that are approved and others that are currently in clinical trials, with an emphasis on human derived natural antibodies with remyleination potential. Our investigative group developed recombinant natural human IgM antibodies against oligodendrocytes and neurons with a potential for CNS repair and remyleination. One such recombinant antibody, rHIgM22 completed a phase 1 clinical trial with no toxicity and with an objective of promoting remyleination in multiple sclerosis. Inclusion of these drugs as a multifaceted approach may further enhance the efficacy of neurorehabilitation in neuroinflammatory and neurodegenerative disorders.

7.
Curr Pharm Des ; 22(40): 6134-6141, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27634184

RESUMO

BACKGROUND: Alterations in gut microbiota, coupled to increased gut permeability are now widely recognized as having a role in the etiology, course and treatment of many medical conditions, including autoimmune and neurodegenerative disorders. METHODS: In this review, the role that such gut changes play over the course of multiple sclerosis (MS) is detailed. RESULTS: Given the wide array of biological factors and processes that have been shown to be altered in MS, including changes in the gut, this allows for a better integration of the diverse array of pathophysiological processes linked to MS. Such pathophysiological processes include increases in oxidative and nitrosative stress, pro-inflammatory immune responses, especially T helper (Th)17 cell proliferation and activation, tryptophan catabolites, pain, fatigue and increased levels of depression. By raising levels of immune activation, increased gut permeability and alterations in gut microbiota impact on all of these MS-associated processes. Alterations in the regulation of local melatonergic pathway activation is proposed to be an important hub for such pathophysiological processes in MS, allowing for the increased frequency of depression that may be prodromal in MS, both in the first episode as well as in relapses, to become more intimately associated with the etiology and course of MS. We propose this occurs by decreasing serotonin availability as a precursor for the melatoninergic pathways. CONCLUSION: Changes in the gut are evident in the early stages of MS, including in paediatric MS, and may interact with pro-inflammatory genetic susceptibility genes to drive the biological underpinnings of MS. Such a conceptualization of the biological underpinnings of MS also has treatment implications.


Assuntos
Depressão/metabolismo , Microbioma Gastrointestinal , Melatonina/metabolismo , Esclerose Múltipla/metabolismo , Triptofano/metabolismo , Animais , Microbioma Gastrointestinal/imunologia , Humanos , Esclerose Múltipla/imunologia , Permeabilidade
8.
J Vis Exp ; (112)2016 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-27404858

RESUMO

Antibodies of the IgM isotype are often neglected as potential therapeutics in human trials, animal models of human diseases as well as detecting agents in standard laboratory techniques. In contrast, several human IgMs demonstrated proof of efficacy in cancer models and models of CNS disorders including multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). Reasons for their lack of consideration include difficulties to express, purify and stabilize IgM antibodies, challenge to identify (non-protein) antigens, low affinity binding and fundamental knowledge gaps in carbohydrate and lipid research. This manuscript uses HIgM12 as an example to provide a detailed protocol to detect antigens by Western blotting, immunoprecipitations and immunocytochemistry. HIgM12 targets polysialic acid (PSA) attached to the neural cell adhesion molecule (NCAM). Early postnatal mouse brain tissue from wild type (WT) and NCAM knockout (KO) mice lacking the three major central nervous system (CNS) splice variants NCAM180, 140 and 120 was used to evaluate the importance of NCAM for binding to HIgM12. Further enzymatic digestion of CNS tissue and cultured CNS cells using endoneuraminidases led us to identify PSA as the specific binding epitope for HIgM12.


Assuntos
Especificidade de Anticorpos , Animais , Humanos , Imunoglobulina M , Camundongos , Moléculas de Adesão de Célula Nervosa , Neurônios , Transdução de Sinais
9.
J Neuroinflammation ; 13(1): 94, 2016 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-27126523

RESUMO

BACKGROUND: Whereas demyelination underlies early neurological symptoms in multiple sclerosis (MS), axonal damage is considered critical for permanent chronic deficits. Intracerebral infection of susceptible mouse strains with Theiler's murine encephalomyelitis virus (TMEV) results in chronic induced demyelinating disease (TMEV-IDD) with progressive axonal loss and neurologic dysfunction similar to progressive forms of MS. We previously reported that treatment of chronic TMEV-IDD mice with a neurite outgrowth-promoting natural human antibody, HIgM12, improved brainstem NAA concentrations and preserved functional motor activity. In order to translate this antibody toward clinical trial, we generated a fully human recombinant form of HIgM12, rHIgM12, determined the optimal in vivo dose for functional improvement in TMEV-IDD, and evaluated the functional preservation of descending spinal cord axons by retrograde labeling. FINDINGS: SJL/J mice at 45 to 90 days post infection (dpi) were studied. A single intraperitoneal dose of 0.25 mg/kg of rHIgM12 per mouse is sufficient to preserve motor function in TMEV-IDD. The optimal dose was 10 mg/kg. rHIgM12 treatment protected the functional transport in spinal cord axons and led to 40 % more Fluoro-Gold-labeled brainstem neurons in retrograde transport studies. This suggests that axons are not only present but also functionally competent. rHIgM12-treated mice also contained more mid-thoracic (T6) spinal cord axons than controls. CONCLUSIONS: This study confirms that a fully human recombinant neurite outgrowth-promoting monoclonal IgM is therapeutic in a model of progressive MS using multiple reparative readouts. The minimum effective dose is similar to that of a remyelination-promoting monoclonal human IgM discovered by our group that is presently in clinical trials for MS.


Assuntos
Anticorpos Monoclonais/farmacologia , Axônios/efeitos dos fármacos , Imunoglobulina M/farmacologia , Esclerose Múltipla/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Axônios/patologia , Tronco Encefálico/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Medula Espinal/patologia , Theilovirus
10.
J Immunol ; 196(10): 4075-81, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27067006

RESUMO

Renal transplant is the treatment of choice for patients with terminal end-stage renal disease. We have previously identified low levels of catalytic IgG as a potential prognosis marker for chronic allograft rejection. The origin and physiopathological relevance of catalytic Abs is not well understood, owing to the fact that catalytic Abs have been studied in relatively small cohorts of patients with rare diseases and/or without systematic follow-up. In the current study, we have followed the evolution of the levels of catalytic IgG in a large cohort of renal transplant patients over a 2-y period. Our results demonstrate that, prior to transplant, patients with renal failure present with heterogeneous levels of IgG hydrolyzing the generic proline-phenylalanine-arginine-methylcoumarinamide (PFR-MCA) substrate. PFR-MCA hydrolysis was greater for patients' IgG than for a therapeutic preparation of pooled IgG from healthy donors. Renal transplant was marked by a drastic decrease in levels of catalytic IgG over 3 mo followed by a steady increase during the next 21 mo. Patients who displayed high levels of catalytic IgG pretransplant recovered high levels of catalytic Abs 2 y posttransplant. Interestingly, IgG-mediated hydrolysis of a model protein substrate, procoagulant factor VIII, did not correlate with that of PFR-MCA prior transplantation, whereas it did 12 mo posttransplant. Taken together, our results suggest that the level of circulating catalytic IgG under pathological conditions is an intrinsic property of each individual's immune system and that recovery of pretransplant levels of catalytic IgG is accompanied by changes in the repertoire of target Ags.


Assuntos
Biomarcadores/metabolismo , Rejeição de Enxerto/imunologia , Sistema Imunitário , Imunoglobulina G/metabolismo , Transplante de Rim , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Catalíticos , Autoanticorpos/metabolismo , Coagulação Sanguínea , Doença Crônica , Fator VIII/metabolismo , Feminino , Seguimentos , Rejeição de Enxerto/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Transplantados , Adulto Jovem
11.
Curr Pharm Des ; 22(8): 1049-59, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26899126

RESUMO

Accumulating evidence demonstrates involvement of tryptophan metabolites and in particular activation of the kynurenine pathway (KP) in neurocognitive disorders under CNS inflammatory conditions. The KP is involved in several brain-associated disorders including Parkinson's disease, AIDS dementia, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, schizophrenia, and brain tumors. Our review is an attempt to address any relevant association between dysregulation of KP and multiple sclerosis (MS), an inflammatory CNS disorder that ultimately leads to demyelinated brain areas and severe neurological deficits. Modulation of KP is a new topic for the field of MS and warrants further research. The availability of potential KP modulators approved for MS may shed some light into the therapeutic potential of KP antagonists for the treatment of MS patients.


Assuntos
Esclerose Múltipla/fisiopatologia , Triptofano/metabolismo , Animais , Progressão da Doença , Humanos
12.
Expert Opin Biol Ther ; 16(6): 827-839, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26914737

RESUMO

INTRODUCTION: Multiple sclerosis (MS) is the most common chronic inflammatory, demyelinating disease of the CNS and results in neurological disability. Existing immunomodulatory and immunosuppressive approaches lower the number of relapses but do not cure or reverse existing deficits nor improve long-term disability in MS patients. AREAS COVERED: Monogenic antibodies were described as treatment options for MS, however the immunogenicity of mouse antibodies hampered the efficacy of potential therapeutics in humans. Availability of improved antibody production technologies resulted in a paradigm shift in MS treatment strategies. In this review, an overview of immunotherapies for MS that use conventional monoclonal antibodies reactive to immune system and their properties and mechanisms of action will be discussed, including recent advances in MS therapeutics and highlight natural autoantibodies (NAbs) that directly target CNS cells. EXPERT OPINION: Recent challenges for MS therapy are the identification of relevant molecular and cellular targets, time frame of treatment, and antibody toxicity profiles to identify safe treatment options for MS patients. The application of monoclonal antibody therapies with better biological efficacy associated with minimum side effects possesses huge clinical potential. Advances in monoclonal antibody technologies that directly target cells of nervous system may promote the CNS regeneration field from bench to bedside.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia/métodos , Esclerose Múltipla/tratamento farmacológico , Animais , Anticorpos Monoclonais/farmacologia , Formação de Anticorpos/efeitos dos fármacos , Formação de Anticorpos/imunologia , Humanos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Imunoterapia/tendências , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/imunologia
13.
Mol Neurobiol ; 53(8): 5217-28, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26409478

RESUMO

Demyelination underlies early neurological symptoms in multiple sclerosis (MS); however, axonal damage is considered critical for permanent chronic deficits. The precise mechanisms by which axonal injury occurs in MS are unclear; one hypothesis is the absence or failure of remyelination, suggesting that promoting remyelination may protect axons from death. This report provides direct evidence that promoting oligodendrocyte remyelination protects axons and maintains transport function. Persistent Theiler's virus infection of Swiss Jim Lambert (SJL)/J mice was used as a model of MS to assess the effects of remyelination on axonal injury following demyelination in the spinal cord. Remyelination was induced using an oligodendrocyte/myelin-specific recombinant human monoclonal IgM, rHIgM22. The antibody is endowed with strong anti-apoptotic and pro-proliferative effects on oligodendrocyte progenitor cells. We used (1)H-magnetic resonance spectroscopy (MRS) at the brainstem to measure N-acetyl-aspartate (NAA) as a surrogate of neuronal health and spinal cord integrity. We found increased brainstem NAA concentrations at 5 weeks post-treatment with rHIgM22, which remained stable out to 10 weeks. Detailed spinal cord morphology studies revealed enhanced remyelination in the rHIgM22-treated group but not in the isotype control antibody- or saline-treated groups. Importantly, we found rHIgM22-mediated remyelination protected small- and medium-caliber mid-thoracic spinal cord axons from damage despite similar demyelination and inflammation across all experimental groups. The most direct confirmation of remyelination-mediated protection of descending neurons was an improvement in retrograde transport. Treatment with rHIgM22 significantly increased the number of retrograde-labeled neurons in the brainstem, indicating that preserved axons are functionally competent. This is direct validation that remyelination preserves spinal cord axons and protects functional axon integrity.


Assuntos
Anticorpos/farmacologia , Axônios/metabolismo , Doenças Desmielinizantes/patologia , Oligodendroglia/metabolismo , Remielinização/efeitos dos fármacos , Animais , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Axônios/efeitos dos fármacos , Axônios/patologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Camundongos , Oligodendroglia/efeitos dos fármacos , Medula Espinal/patologia , Theilovirus/efeitos dos fármacos , Theilovirus/fisiologia
14.
Curr Pharm Des ; 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26667001

RESUMO

Accumulating evidence demonstrates involvement of tryptophan metabolites and in particular activation of the kynurenine pathway (KP) in neurocognitive disorders under CNS inflammatory conditions. The KP is involved in several brain-associated disorders including Parkinson's disease, AIDS dementia, Alzheimer's disease, Huntington's disease, amyotrophic lateral sclerosis, schizophrenia, and brain tumors. Our review is an attempt to address any relevant association between dysregulation of KP and multiple sclerosis (MS), an inflammatory CNS disorder that ultimately leads to demyelinated brain areas and severe neurological deficits. Modulation of KP is a new topic for the field of MS and warrants further research. The availability of potential KP modulators approved for MS may shed some light into the therapeutic potential of KP antagonists for the treatment of MS patients.

15.
JAMA Neurol ; 72(11): 1346-53, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26389734

RESUMO

IMPORTANCE: Modulating the immune system does not reverse long-term disability in neurologic disorders. Better neuroregenerative and neuroprotective treatment strategies are needed for neuroinflammatory and neurodegenerative diseases. OBJECTIVE: To review the role of monoclonal, naturally occurring antibodies (NAbs) as novel therapeutic molecules for treatment of neurologic disorders. EVIDENCE REVIEW: Peer-reviewed articles, including case reports, case series, retrospective reviews, prospective randomized clinical trials, and basic science reports, were identified in a PubMed search for articles about NAbs and neurologic disorders that were published from January 1, 1964, through June 30, 2015. We concentrated our review on multiple sclerosis, Parkinson disease, Alzheimer disease, and amyotrophic lateral sclerosis. FINDINGS: Many insults, including trauma, ischemia, infection, inflammation, and neurodegeneration, result in irreversible damage to the central nervous system. Central nervous system injury often results in a pervasive inhibitory microenvironment that hinders regeneration. A common targeted drug development strategy is to identify molecules with high potency in animal models. Many approaches often fail in the clinical setting owing to a lack of efficacy in human diseases (eg, less than the response demonstrated in animal models) or a high incidence of toxic effects. An alternative approach is to identify NAbs in humans because these therapeutic molecules have potential physiologic function without toxic effects. NAbs of the IgG, IgA, or IgM isotype contain germline or close to germline sequences and are reactive to self-components, altered self-components, or foreign antigens. Our investigative group developed recombinant, autoreactive, natural human IgM antibodies directed against oligodendrocytes or neurons with therapeutic potential for central nervous system repair. One such molecule, recombinant HIgM22, directed against myelin and oligodendrocytes completed a successful phase 1 clinical trial without toxic effects with the goal of promoting remyelination in multiple sclerosis. CONCLUSIONS AND RELEVANCE: Animal studies demonstrate that certain monoclonal NAbs are beneficial as therapeutic agents for neurologic diseases. This class of antibodies represents a unique source from which to develop a new class of disease-modifying therapies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Isotipos de Imunoglobulinas/imunologia , Doenças do Sistema Nervoso/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Animais , Humanos , Doenças do Sistema Nervoso/imunologia
16.
Dis Model Mech ; 8(8): 831-42, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26035393

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating, fatal neurological disease that primarily affects spinal cord anterior horn cells and their axons for which there is no treatment. Here we report the use of a recombinant natural human IgM that binds to the surface of neurons and supports neurite extension, rHIgM12, as a therapeutic strategy in murine models of human ALS. A single 200 µg intraperitoneal dose of rHIgM12 increases survival in two independent genetic-based mutant SOD1 mouse strains (SOD1G86R and SOD1G93A) by 8 and 10 days, delays the onset of neurological deficits by 16 days, delays the onset of weight loss by 5 days, and preserves spinal cord axons and anterior horn neurons. Immuno-overlay of thin layer chromatography and surface plasmon resonance show that rHIgM12 binds with high affinity to the complex gangliosides GD1a and GT1b. Addition of rHIgM12 to neurons in culture increases α-tubulin tyrosination levels, suggesting an alteration of microtubule dynamics. We previously reported that a single peripheral dose of rHIgM12 preserved neurological function in a murine model of demyelination with axon loss. Because rHIgM12 improves three different models of neurological disease, we propose that the IgM might act late in the cascade of neuronal stress and/or death by a broad mechanism.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Gangliosídeos/metabolismo , Imunoglobulina M/uso terapêutico , Esclerose Lateral Amiotrófica/imunologia , Esclerose Lateral Amiotrófica/patologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta Imunológica , Epitopos/química , Gangliosídeos/química , Humanos , Bicamadas Lipídicas/metabolismo , Camundongos , Camundongos Transgênicos , Microtúbulos/metabolismo , Modelos Moleculares , Neuritos/metabolismo , Neuritos/patologia , Ligação Proteica , Proteínas Recombinantes/uso terapêutico , Solubilidade , Medula Espinal/patologia , Superóxido Dismutase/metabolismo , Ressonância de Plasmônio de Superfície , Análise de Sobrevida , Tubulina (Proteína)/metabolismo
17.
PLoS One ; 10(5): e0128007, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26020269

RESUMO

Neonatal white matter injury (nWMI) is an increasingly common cause of cerebral palsy that results predominantly from hypoxic injury to progenitor cells including those of the oligodendrocyte lineage. Existing mouse models of nWMI utilize prolonged periods of hypoxia during the neonatal period, require complex cross-fostering and exhibit poor growth and high mortality rates. Abnormal CNS myelin composition serves as the major explanation for persistent neuro-motor deficits. Here we developed a simplified model of nWMI with low mortality rates and improved growth without cross-fostering. Neonatal mice are exposed to low oxygen from postnatal day (P) 3 to P7, which roughly corresponds to the period of human brain development between gestational weeks 32 and 36. CNS hypomyelination is detectable for 2-3 weeks post injury and strongly correlates with levels of body and brain weight loss. Immediately following hypoxia treatment, cell death was evident in multiple brain regions, most notably in superficial and deep cortical layers as well as the subventricular zone progenitor compartment. PDGFαR, Nkx2.2, and Olig2 positive oligodendrocyte progenitor cell were significantly reduced until postnatal day 27. In addition to CNS dysmyelination we identified a novel pathological marker for adult hypoxic animals that strongly correlates with life-long neuro-motor deficits. Mice reared under hypoxia reveal an abnormal spinal neuron composition with increased small and medium diameter axons and decreased large diameter axons in thoracic lateral and anterior funiculi. Differences were particularly pronounced in white matter motor tracts left and right of the anterior median fissure. Our findings suggest that 4 days of exposure to hypoxia are sufficient to induce experimental nWMI in CD1 mice, thus providing a model to test new therapeutics. Pathological hallmarks of this model include early cell death, decreased OPCs and hypomyelination in early postnatal life, followed by dysmyelination, abnormal spinal neuron composition, and neuro-motor deficits in adulthood.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Neurônios Motores/metabolismo , Células-Tronco Neurais/metabolismo , Substância Branca/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio , Humanos , Hipóxia-Isquemia Encefálica/patologia , Camundongos , Neurônios Motores/patologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Proteínas do Tecido Nervoso/biossíntese , Células-Tronco Neurais/patologia , Proteínas Nucleares , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fatores de Transcrição , Substância Branca/patologia
18.
J Neuroinflammation ; 12: 83, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25924771

RESUMO

BACKGROUND: Intracerebral infection of susceptible mouse strains with Theiler's murine encephalomyelitis virus (TMEV) results in chronic demyelinating disease with progressive axonal loss and neurologic dysfunction similar to progressive forms of multiple sclerosis (MS). We previously showed that as the disease progresses, a marked decrease in brainstem N-acetyl aspartate (NAA; metabolite associated with neuronal integrity) concentrations, reflecting axon health, is measured. We also demonstrated stimulation of neurite outgrowth by a neuron-binding natural human antibody, IgM12. Treatment with either the serum-derived or recombinant human immunoglobulin M 12 (HIgM12) preserved functional motor activity in the TMEV model. In this study, we examined IgM-mediated changes in brainstem NAA concentrations and central nervous system (CNS) pathology. FINDINGS: (1)H-magnetic resonance spectroscopy (MRS) showed that treatment with HIgM12 significantly increased brainstem NAA concentrations compared to controls in TMEV-infected mice. Pathologic analysis demonstrated a significant preservation of axons in the spinal cord of animals treated with HIgM12. CONCLUSIONS: This study links drug efficacy of slowing deficits with axon preservation and NAA concentrations in the brainstem in a model of progressive MS. HIgM12-mediated changes of NAA concentrations in the brainstem are a surrogate marker of axon injury/preservation throughout the spinal cord. This study provides proof-of-concept that a neuron-reactive human IgM can be therapeutic and provides a biomarker for clinical trials.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Ácido Aspártico/análogos & derivados , Tronco Encefálico/metabolismo , Glicoproteínas de Membrana/imunologia , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Proteínas do Envelope Viral/imunologia , Animais , Ácido Aspártico/metabolismo , Axônios/efeitos dos fármacos , Encéfalo/patologia , Tronco Encefálico/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Esclerose Múltipla/etiologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Estatísticas não Paramétricas , Theilovirus/imunologia
19.
J Neurochem ; 134(5): 865-78, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25866077

RESUMO

CNS regeneration is a desirable goal for diseases of brain and spinal cord. Current therapeutic strategies for the treatment of multiple sclerosis (MS) aim to eliminate detrimental effects of the immune system, so far without reversing disability or affecting long-term prognosis in patients. Approachable molecular targets that stimulate CNS repair are not part of the clinical praxis or have not been identified yet. The purpose of this study was to identify the molecular target of the human monoclonal antibody HIgM12. HIgM12 reverses motor deficits in chronically demyelinated mice, a model of MS. Here, we identified polysialic acid (PSA) attached to the neural cell adhesion molecule (NCAM) as the antigen for HIgM12 by using different NCAM knockout strains and through PSA removal from the NCAM protein core. Antibody binding to CNS tissue and primary cells, antibody-mediated cell adhesion, and neurite outgrowth on HIgM12-coated nitrocellulose was detected only in the presence of PSA as assessed by western blotting, immunoprecipitation, immunocytochemistry, and histochemistry. We conclude that HIgM12 mediates its in vivo and in vitro effects through binding to PSA and has the potential to be an effective therapy for MS and neurodegenerative diseases. The human antibody HIgM12 stimulates neurite outgrowth in vitro and promotes function in chronically demyelinated mice, a model of multiple sclerosis. The cellular antigen for HIgM12 was undetermined. Here, we identified polysialic acid attached to NCAM (neural cell adhesion molecule) as the cellular target for HIgM12. This includes glial fibrillary acidic protein (GFAP)-positive mouse astrocytes (GFAP, red; HIgM12, green; DAPI, blue) among other cell types of the central nervous system. These findings indicate a new strategy for the treatment of neuro-motor disorders including multiple sclerosis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígenos/imunologia , Antígeno CD56/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/tratamento farmacológico , Esclerose Múltipla/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Ácidos Siálicos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Reações Antígeno-Anticorpo , Antígeno CD56/química , Antígeno CD56/genética , Adesão Celular , Células Cultivadas , Cerebelo/citologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Glicosilação/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/imunologia , Regeneração Nervosa , Neuraminidase/farmacologia , Neuritos/efeitos dos fármacos , Doenças Neurodegenerativas/imunologia , Neuroglia/citologia , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley
20.
Nucleic Acid Ther ; 25(1): 11-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25536292

RESUMO

DNA aptamer oligonucleotides and their protein conjugates show promise as therapeutics in animal models of diseases such as multiple sclerosis. These molecules are large and highly charged, raising questions about their biodistribution and pharmacokinetics in mammals. Here we exploit the power of quantitative polymerase chain reaction to accurately quantitate the tissue distribution of 40-nucleotide DNA aptamers and their streptavidin conjugates after intraperitoneal injection in mice. We show remarkably rapid distribution to peripheral tissues including the central nervous system. Modeling of tissue distribution data reveals the importance of DNA aptamer sequence, 3' modification, and protein conjugation in enhancing tissue exposure. These data help to interpret the previously observed effectiveness of aptamer conjugates, as opposed to free aptamers, in stimulating central nervous system remyelination in a mouse model of multiple sclerosis.


Assuntos
Aptâmeros de Nucleotídeos/farmacocinética , Animais , Animais não Endogâmicos , Aptâmeros de Nucleotídeos/genética , Área Sob a Curva , Sequência de Bases , Feminino , Terapia Genética , Camundongos , Esclerose Múltipla/terapia , Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...