Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Front Immunol ; 14: 1310094, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38098484

RESUMO

Midkine (MDK) is a neurotrophic growth factor highly expressed during embryogenesis with important functions related to growth, proliferation, survival, migration, angiogenesis, reproduction, and repair. Recent research has indicated that MDK functions as a key player in autoimmune disorders of the central nervous system (CNS), such as Multiple Sclerosis (MS) and is a promising therapeutic target for the treatment of brain tumors, acute injuries, and other CNS disorders. This review summarizes the modes of action and immunological functions of MDK both in the peripheral immune compartment and in the CNS, particularly in the context of traumatic brain injury, brain tumors, neuroinflammation, and neurodegeneration. Moreover, we discuss the role of MDK as a central mediator of neuro-immune crosstalk, focusing on the interactions between CNS-infiltrating and -resident cells such as astrocytes, microglia, and oligodendrocytes. Finally, we highlight the therapeutic potential of MDK and discuss potential therapeutic approaches for the treatment of neurological disorders.


Assuntos
Lesões Encefálicas Traumáticas , Neoplasias Encefálicas , Humanos , Midkina , Sistema Nervoso Central/patologia , Lesões Encefálicas Traumáticas/patologia , Astrócitos/patologia , Neoplasias Encefálicas/patologia
2.
Brain Commun ; 5(4): fcad206, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37564830

RESUMO

The programmed cell death protein 1/programmed cell death ligand 1 axis plays an important role in the adaptive immune system and has influence on neoplastic and inflammatory diseases, while its role in multiple sclerosis is unclear. Here, we aimed to analyse expression patterns of programmed cell death protein 1 and programmed cell death ligand 1 on peripheral blood mononuclear cells and their soluble variants in multiple sclerosis patients and controls, to determine their correlation with clinical disability and disease activity. In a cross-sectional study, we performed in-depth flow cytometric immunophenotyping of peripheral blood mononuclear cells and analysed soluble programmed cell death protein 1 and programmed cell death ligand 1 serum levels in patients with relapsing-remitting multiple sclerosis and controls. In comparison to control subjects, relapsing-remitting multiple sclerosis patients displayed distinct cellular programmed cell death protein 1/programmed cell death ligand 1 expression patterns in immune cell subsets and increased soluble programmed cell death ligand 1 levels, which correlated with clinical measures of disability and MRI activity over time. This study extends our knowledge of how programmed cell death protein 1 and programmed cell death ligand 1 are expressed in the membranes of patients with relapsing-remitting multiple sclerosis and describes for the first time the elevation of soluble programmed cell death ligand 1 in the blood of multiple sclerosis patients. The distinct expression pattern of membrane-bound programmed cell death protein 1 and programmed cell death ligand 1 and the correlation between soluble programmed cell death ligand 1, membrane-bound programmed cell death ligand 1, disease and clinical factors may offer therapeutic potential in the setting of multiple sclerosis and might improve future diagnosis and clinical decision-making.

3.
Science ; 379(6636): 1023-1030, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36893254

RESUMO

Cell-cell interactions in the central nervous system play important roles in neurologic diseases. However, little is known about the specific molecular pathways involved, and methods for their systematic identification are limited. Here, we developed a forward genetic screening platform that combines CRISPR-Cas9 perturbations, cell coculture in picoliter droplets, and microfluidic-based fluorescence-activated droplet sorting to identify mechanisms of cell-cell communication. We used SPEAC-seq (systematic perturbation of encapsulated associated cells followed by sequencing), in combination with in vivo genetic perturbations, to identify microglia-produced amphiregulin as a suppressor of disease-promoting astrocyte responses in multiple sclerosis preclinical models and clinical samples. Thus, SPEAC-seq enables the high-throughput systematic identification of cell-cell communication mechanisms.


Assuntos
Anfirregulina , Astrócitos , Comunicação Autócrina , Testes Genéticos , Técnicas Analíticas Microfluídicas , Microglia , Astrócitos/fisiologia , Testes Genéticos/métodos , Ensaios de Triagem em Larga Escala , Técnicas Analíticas Microfluídicas/métodos , Microglia/fisiologia , Anfirregulina/genética , Comunicação Autócrina/genética , Expressão Gênica , Humanos
4.
J Mol Med (Berl) ; 99(5): 663-671, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33398468

RESUMO

Mesenchymal stem cells (MSCs) are promising candidates for the development of cell-based drug delivery systems for autoimmune inflammatory diseases, such as multiple sclerosis (MS). Here, we investigated the effect of Ro-31-8425, an ATP-competitive kinase inhibitor, on the therapeutic properties of MSCs. Upon a simple pretreatment procedure, MSCs spontaneously took up and then gradually released significant amounts of Ro-31-8425. Ro-31-8425 (free or released by MSCs) suppressed the proliferation of CD4+ T cells in vitro following polyclonal and antigen-specific stimulation. Systemic administration of Ro-31-8425-loaded MSCs ameliorated the clinical course of experimental autoimmune encephalomyelitis (EAE), a murine model of MS, displaying a stronger suppressive effect on EAE than control MSCs or free Ro-31-8425. Ro-31-8425-MSC administration resulted in sustained levels of Ro-31-8425 in the serum of EAE mice, modulating immune cell trafficking and the autoimmune response during EAE. Collectively, these results identify MSC-based drug delivery as a potential therapeutic strategy for the treatment of autoimmune diseases. KEY MESSAGES: MSCs can spontaneously take up the ATP-competitive kinase inhibitor Ro-31-8425. Ro-31-8425-loaded MSCs gradually release Ro-31-8425 and exhibit sustained suppression of T cells. Ro-31-8425-loaded MSCs have more sustained serum levels of Ro-31-8425 than free Ro-31-8425. Ro-31-8425-loaded MSCs are more effective than MSCs and free Ro-31-8425 for EAE therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Indóis/administração & dosagem , Maleimidas/administração & dosagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Esclerose Múltipla/tratamento farmacológico , Transplante Heterólogo/métodos , Animais , Proliferação de Células/efeitos dos fármacos , Liberação Controlada de Fármacos , Encefalomielite Autoimune Experimental/sangue , Encefalomielite Autoimune Experimental/imunologia , Inibidores Enzimáticos/sangue , Feminino , Humanos , Imunidade/efeitos dos fármacos , Indóis/sangue , Maleimidas/sangue , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/sangue , Esclerose Múltipla/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Distribuição Tecidual , Resultado do Tratamento
5.
Nat Neurosci ; 23(8): 939-951, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32690969

RESUMO

Zika virus (ZIKV) is a flavivirus linked to multiple birth defects including microcephaly, known as congenital ZIKV syndrome. The identification of host factors involved in ZIKV replication may guide efficacious therapeutic interventions. In genome-wide transcriptional studies, we found that ZIKV infection triggers aryl hydrocarbon receptor (AHR) activation. Specifically, ZIKV infection induces kynurenine (Kyn) production, which activates AHR, limiting the production of type I interferons (IFN-I) involved in antiviral immunity. Moreover, ZIKV-triggered AHR activation suppresses intrinsic immunity driven by the promyelocytic leukemia (PML) protein, which limits ZIKV replication. AHR inhibition suppressed the replication of multiple ZIKV strains in vitro and also suppressed replication of the related flavivirus dengue. Finally, AHR inhibition with a nanoparticle-delivered AHR antagonist or an inhibitor developed for human use limited ZIKV replication and ameliorated newborn microcephaly in a murine model. In summary, we identified AHR as a host factor for ZIKV replication and PML protein as a driver of anti-ZIKV intrinsic immunity.


Assuntos
Receptores de Hidrocarboneto Arílico/metabolismo , Replicação Viral , Zika virus/metabolismo , Animais , Chlorocebus aethiops , Células Hep G2 , Humanos , Células Vero , Infecção por Zika virus/metabolismo
6.
Nature ; 578(7796): 593-599, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32051591

RESUMO

Multiple sclerosis is a chronic inflammatory disease of the CNS1. Astrocytes contribute to the pathogenesis of multiple sclerosis2, but little is known about the heterogeneity of astrocytes and its regulation. Here we report the analysis of astrocytes in multiple sclerosis and its preclinical model experimental autoimmune encephalomyelitis (EAE) by single-cell RNA sequencing in combination with cell-specific Ribotag RNA profiling, assay for transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing (ChIP-seq), genome-wide analysis of DNA methylation and in vivo CRISPR-Cas9-based genetic perturbations. We identified astrocytes in EAE and multiple sclerosis that were characterized by decreased expression of NRF2 and increased expression of MAFG, which cooperates with MAT2α to promote DNA methylation and represses antioxidant and anti-inflammatory transcriptional programs. Granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in astrocytes drives the expression of MAFG and MAT2α and pro-inflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, multiple sclerosis. Our results identify candidate therapeutic targets in multiple sclerosis.


Assuntos
Astrócitos/patologia , Sistema Nervoso Central/patologia , Inflamação/patologia , Fator de Transcrição MafG/genética , Proteínas Repressoras/genética , Animais , Antioxidantes/metabolismo , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Metilação de DNA , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação/genética , Masculino , Metionina Adenosiltransferase/genética , Camundongos , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Fator 2 Relacionado a NF-E2/genética , Análise de Sequência de RNA , Transdução de Sinais , Transcrição Gênica
8.
Nat Neurosci ; 22(5): 729-740, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30962630

RESUMO

Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.


Assuntos
Antígenos CD/metabolismo , Apirase/metabolismo , Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Cinurenina/metabolismo , Macrófagos/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Glioblastoma/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Receptores de Lipopolissacarídeos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Fator de Transcrição STAT1 , Fator de Transcrição STAT3/metabolismo , Linfócitos T/imunologia , Microambiente Tumoral
9.
Nat Rev Immunol ; 19(3): 184-197, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30718831

RESUMO

The environment, diet, microbiota and body's metabolism shape complex biological processes in health and disease. However, our understanding of the molecular pathways involved in these processes is still limited. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that integrates environmental, dietary, microbial and metabolic cues to control complex transcriptional programmes in a ligand-specific, cell-type-specific and context-specific manner. In this Review, we summarize our current knowledge of AHR and the transcriptional programmes it controls in the immune system. Finally, we discuss the role of AHR in autoimmune and neoplastic diseases of the central nervous system, with a special focus on the gut immune system, the gut-brain axis and the therapeutic potential of targeting AHR in neurological disorders.


Assuntos
Imunidade Adaptativa/imunologia , Doenças Autoimunes do Sistema Nervoso/imunologia , Encéfalo/imunologia , Regulação da Expressão Gênica/imunologia , Intestinos/imunologia , Receptores de Hidrocarboneto Arílico/imunologia , Imunidade Adaptativa/genética , Animais , Doenças Autoimunes do Sistema Nervoso/genética , Dieta , Meio Ambiente , Microbioma Gastrointestinal/imunologia , Humanos , Microbiota/imunologia , Terapia de Alvo Molecular , Transdução de Sinais
10.
Nature ; 557(7707): 724-728, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769726

RESUMO

Microglia and astrocytes modulate inflammation and neurodegeneration in the central nervous system (CNS)1-3. Microglia modulate pro-inflammatory and neurotoxic activities in astrocytes, but the mechanisms involved are not completely understood4,5. Here we report that TGFα and VEGF-B produced by microglia regulate the pathogenic activities of astrocytes in the experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis. Microglia-derived TGFα acts via the ErbB1 receptor in astrocytes to limit their pathogenic activities and EAE development. Conversely, microglial VEGF-B triggers FLT-1 signalling in astrocytes and worsens EAE. VEGF-B and TGFα also participate in the microglial control of human astrocytes. Furthermore, expression of TGFα and VEGF-B in CD14+ cells correlates with the multiple sclerosis lesion stage. Finally, metabolites of dietary tryptophan produced by the commensal flora control microglial activation and TGFα and VEGF-B production, modulating the transcriptional program of astrocytes and CNS inflammation through a mechanism mediated by the aryl hydrocarbon receptor. In summary, we identified positive and negative regulators that mediate the microglial control of astrocytes. Moreover, these findings define a pathway through which microbial metabolites limit pathogenic activities of microglia and astrocytes, and suppress CNS inflammation. This pathway may guide new therapies for multiple sclerosis and other neurological disorders.


Assuntos
Astrócitos/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/microbiologia , Microglia/metabolismo , Animais , Astrócitos/patologia , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/microbiologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/prevenção & controle , Receptores ErbB/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Inflamação/prevenção & controle , Receptores de Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/patologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Receptores de Hidrocarboneto Arílico/metabolismo , Simbiose , Fator de Crescimento Transformador alfa/biossíntese , Fator de Crescimento Transformador alfa/metabolismo , Triptofano/deficiência , Triptofano/metabolismo , Fator B de Crescimento do Endotélio Vascular/biossíntese , Fator B de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
11.
Sci Rep ; 8(1): 4970, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29563571

RESUMO

The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor with important functions in the immune response and cancer. AHR agonists are provided by the environment, the commensal flora and the metabolism. Considering AHR physiological functions, AHR agonists may have important effects on health and disease. Thus, the quantification of AHR agonists in biological samples is of scientific and clinical relevance. We compared different reporter systems for the detection of AHR agonists in serum samples of Multiple Sclerosis (MS) patients, and assessed the influence of transfection methods and cell lines in a reporter-based in vitro assay. While the use of stable or transient reporters did not influence the measurement of AHR agonistic activity, the species of the cell lines used in these reporter assays had important effects on the reporter readings. These observations suggest that cell-specific factors influence AHR activation and signaling. Thus, based on the reported species selectivity of AHR ligands and the cell species-of-origin effects that we describe in this manuscript, the use of human cell lines is encouraged for the analysis of AHR agonistic activity in human samples. These findings may be relevant for the analysis of AHR agonists in human samples in the context of inflammatory and neoplastic disorders.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Bioensaio/métodos , Genes Reporter , Esclerose Múltipla/sangue , Receptores de Hidrocarboneto Arílico/agonistas , Adulto , Idoso , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Poluentes Ambientais/sangue , Feminino , Células HEK293 , Células Hep G2 , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Receptores de Hidrocarboneto Arílico/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Transfecção
12.
Proc Natl Acad Sci U S A ; 114(8): 2012-2017, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28167760

RESUMO

Multiple sclerosis (MS) is an autoimmune inflammatory demyelinating disease of the CNS that causes disability in young adults as a result of the irreversible accumulation of neurological deficits. Although there are potent disease-modifying agents for its initial relapsing-remitting phase, these therapies show limited efficacy in secondary progressive MS (SPMS). Thus, there is an unmet clinical need for the identification of disease mechanisms and potential therapeutic approaches for SPMS. Here, we show that the sphingosine 1-phosphate receptor (S1PR) modulator fingolimod (FTY720) ameliorated chronic progressive experimental autoimmune encephalomyelitis in nonobese diabetic mice, an experimental model that resembles several aspects of SPMS, including neurodegeneration and disease progression driven by the innate immune response in the CNS. Indeed, S1PR modulation by FTY720 in murine and human astrocytes suppressed neurodegeneration-promoting mechanisms mediated by astrocytes, microglia, and CNS-infiltrating proinflammatory monocytes. Genome-wide studies showed that FTY720 suppresses transcriptional programs associated with the promotion of disease progression by astrocytes. The study of the molecular mechanisms controlling these transcriptional modules may open new avenues for the development of therapeutic strategies for progressive MS.


Assuntos
Astrócitos/efeitos dos fármacos , Imunossupressores/farmacologia , Esclerose Múltipla Crônica Progressiva/tratamento farmacológico , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Astrócitos/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Feminino , Cloridrato de Fingolimode/farmacologia , Cloridrato de Fingolimode/uso terapêutico , Humanos , Imunossupressores/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Microglia/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Esclerose Múltipla Crônica Progressiva/patologia , Cultura Primária de Células , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Transcriptoma/efeitos dos fármacos
13.
Acta Neuropathol Commun ; 2: 27, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24606807

RESUMO

INTRODUCTION: Natalizumab blocks α4-integrins and is a prototypic agent for a series of anti-inflammatory drugs that impair trafficking of immune cells into the CNS. However, modulation of the access of immune cells to the CNS is associated with impaired immune surveillance and detrimental viral infections of the CNS. Here, we explored the potency of cellular immune responses within the CNS to protect against viral encephalitis in mice with T cell conditional disruption of VLA-4 integrin (α4ß1) expression. RESULTS: While VLA-4 expression in virus specific Th1 cells is non-redundant for their ability to access the CNS, α4-integrin deficient Th17 cells enter the CNS compartment and generate an inflammatory milieu upon intrathecal vaccinia virus (VV) infection. However, in contrast to Th1 cells that can adopt direct cytotoxic properties, Th17 cells fail to clear the virus due to insufficient Eomes induced perforin-1 expression. CONCLUSION: The quality of the intrathecal cellular antiviral response under conditions of impaired VLA-4 function jeopardizes host protection. Our functional in vivo data extend our mechanistic understanding of anti-viral immunity in the CNS and help to estimate the risk potential of upcoming therapeutic agents that target the trafficking of immune cells into distinct anatomical compartments.


Assuntos
Transferência Adotiva/métodos , Integrina alfa4beta1/imunologia , Meningoencefalite/imunologia , Meningoencefalite/prevenção & controle , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos/uso terapêutico , Antígenos CD4/genética , Diferenciação Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/genética , Adjuvante de Freund/toxicidade , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/imunologia , Proteínas de Homeodomínio/genética , Integrina alfa4beta1/genética , Meningoencefalite/complicações , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Perforina/genética , Perforina/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Infecções por Retroviridae/complicações
14.
Cytotherapy ; 13(7): 802-13, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21506889

RESUMO

BACKGROUND AIMS: Reprogramming of multipotent adult bone marrow (BM)-derived mesenchymal stromal/stem cells (MSC) (BM-MSC) represents one of several strategies for cell-based therapy of diabetes. However, reprogramming primary BM-MSC into pancreatic endocrine lineages has not yet been consistently demonstrated. METHODS: To unravel the role and interaction of key factors governing this process, we used well-characterized telomerase-immortalized human MSC (hMSC-TERT). Pancreatic endocrine differentiation in hMSC-TERT was induced by two major in vitro strategies: (i) endocrine-promoting culture conditions and (ii) ectopic expression of two master regulatory genes of the endocrine lineage, human neurogenin 3 (NGN3) and human pancreatic duodenal homeobox 1 (PDX1). RESULTS: Both approaches triggered pancreatic endocrine gene expression, notably insulin, glucose-transporter 2 and somatostatin. Transgenic overexpression of NGN3 and/or PDX1 proteins not only induced direct target genes, such as NEUROD1 and insulin, and but also triggered parts of the gene expression cascade that is involved in pancreatic endocrine differentiation. Notably, ectopic NGN3 alone was sufficient to initiate the expression of specific beta-cell lineage-associated genes, most importantly PDX1 and insulin. This was demonstrated both transcriptionally by mRNA expression and reporter gene analyzes and at a protein level by Western blotting. Such reprogramming of hMSC-TERT cells induced glucose-insensitive insulin biosynthesis and secretion. CONCLUSIONS: Our results indicate that establishment of glucose-dependent insulin secretion in partially reprogrammed human MSC may depend on additional maturation factors. Moreover, hMSC-TERT provides a suitable cell model for investigating further the molecular mechanisms of reprogramming and maturation of adult MSC towards pancreatic endocrine lineages.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células da Medula Óssea/citologia , Proteínas de Homeodomínio/genética , Ilhotas Pancreáticas/citologia , Células-Tronco Mesenquimais/citologia , Proteínas do Tecido Nervoso/genética , Transativadores/genética , Adulto , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores , Peptídeo C/genética , Peptídeo C/metabolismo , Diferenciação Celular , Linhagem Celular Transformada , Células Cultivadas , Meios de Cultura , Regulação da Expressão Gênica , Transportador de Glucose Tipo 2/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Secreção de Insulina , Masculino , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas , Somatostatina/genética , Telomerase/genética , Telomerase/metabolismo , Transativadores/metabolismo
15.
J Immunol ; 185(10): 6338-47, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20943998

RESUMO

Active immunization with amyloid-ß (Aß) peptide 1-42 reverses amyloid plaque deposition in the CNS of patients with Alzheimer's disease and in amyloid precursor protein transgenic mice. However, this treatment may also cause severe, life-threatening meningoencephalitis. Physiological responses to immunization with Aß(1-42) are poorly understood. In this study, we characterized cognitive and immunological consequences of Aß(1-42)/CFA immunization in C57BL/6 mice. In contrast to mice immunized with myelin oligodendrocyte glycoprotein (MOG)(35-55)/CFA or CFA alone, Aß(1-42)/CFA immunization resulted in impaired exploratory activity, habituation learning, and spatial-learning abilities in the open field. As morphological substrate of this neurocognitive phenotype, we identified a disseminated, nonfocal immune cell infiltrate in the CNS of Aß(1-42)/CFA-immunized animals. In contrast to MOG(35-55)/CFA and PBS/CFA controls, the majority of infiltrating cells in Aß(1-42)/CFA-immunized mice were CD11b(+)CD14(+) and CD45(high), indicating their blood-borne monocyte/macrophage origin. Immunization with Aß(1-42)/CFA was significantly more potent than immunization with MOG(35-55)/CFA or CFA alone in activating macrophages in the secondary lymphoid compartment and peripheral tissues. Studies with TLR2/4-deficient mice revealed that the TLR2/4 pathway mediated the Aß(1-42)-dependent proinflammatory cytokine release from cells of the innate immune system. In line with this, TLR2/4 knockout mice were protected from cognitive impairment upon immunization with Aß(1-42)/CFA. Thus, this study identifies adjuvant effects of Aß(1-42), which result in a clinically relevant neurocognitive phenotype highlighting potential risks of Aß immunotherapy.


Assuntos
Vacinas contra Alzheimer/efeitos adversos , Peptídeos beta-Amiloides/imunologia , Imunidade Inata/imunologia , Transtornos da Memória/imunologia , Fragmentos de Peptídeos/imunologia , Vacinação/efeitos adversos , Adjuvantes Imunológicos/farmacologia , Vacinas contra Alzheimer/imunologia , Peptídeos beta-Amiloides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Separação Celular , Citocinas/biossíntese , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Adjuvante de Freund/imunologia , Adjuvante de Freund/farmacologia , Humanos , Macrófagos/imunologia , Transtornos da Memória/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Fragmentos de Peptídeos/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/imunologia , Receptor 4 Toll-Like/biossíntese , Receptor 4 Toll-Like/imunologia
16.
Am J Physiol Endocrinol Metab ; 291(6): E1168-76, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16822955

RESUMO

p8 protein expression is known to be upregulated in the exocrine pancreas during acute pancreatitis. Own previous work revealed glucose-dependent p8 expression also in endocrine pancreatic beta-cells. Here we demonstrate that glucose-induced INS-1 beta-cell expansion is preceded by p8 protein expression. Moreover, isopropylthiogalactoside (IPTG)-induced p8 overexpression in INS-1 beta-cells (p8-INS-1) enhances cell proliferation and expansion in the presence of glucose only. Although beta-cell-related gene expression (PDX-1, proinsulin I, GLUT2, glucokinase, amylin) and function (insulin content and secretion) are slightly reduced during p8 overexpression, removal of IPTG reverses beta-cell function within 24 h to normal levels. In addition, insulin secretion of p8-INS-1 beta-cells in response to 0-25 mM glucose is not altered by preceding p8-induced beta-cell expansion. Adenovirally transduced p8 overexpression in primary human pancreatic islets increases proliferation, expansion, and cumulative insulin secretion in vitro. Transplantation of mock-transduced control islets under the kidney capsule of immunosuppressed streptozotocin-diabetic mice reduces blood glucose and increases human C-peptide serum concentrations to stable levels after 3 days. In contrast, transplantation of equal numbers of p8-transduced islets results in a continuous decrease of blood glucose and increase of human C-peptide beyond 3 days, indicating p8-induced expansion of transplanted human beta-cells in vivo. This is underlined by a doubling of insulin content in kidneys containing p8-transduced islet grafts explanted on day 9. These results establish p8 as a novel molecular mediator of glucose-induced pancreatic beta-cell expansion in vitro and in vivo and support the notion of existing beta-cell replication in the adult organism.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Glucose/fisiologia , Células Secretoras de Insulina/fisiologia , Proteínas de Neoplasias/fisiologia , Adenoviridae/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Glicemia/metabolismo , Western Blotting , Peso Corporal/fisiologia , Peptídeo C/metabolismo , Contagem de Células , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/cirurgia , Humanos , Insulina/biossíntese , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Transplante das Ilhotas Pancreáticas , Isopropiltiogalactosídeo/farmacologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/biossíntese , Organismos Geneticamente Modificados , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Transplante Heterólogo
17.
Hum Mutat ; 24(4): 351-2, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15365996

RESUMO

Hereditary nonpolyposis colorectal cancer (HNPCC) is one of the most common hereditary cancer-susceptibility syndromes. Germline mutations in mismatch repair genes are associated with the clinical phenotype of HNPCC. We report ten novel germline mutations, three in MSH2 and seven in MLH1. All but one mutation have been found in families fulfilling criteria of the Bethesda guidelines; four of them additionally fulfilled the Amsterdam criteria I or II. Eight mutations were considered pathogenic and predictive diagnostics in healthy family members at risk shall be undertaken; these include five frameshift mutations leading to premature stop codons, in MSH2: c.1672delT (p.S558Xfs) and c.2466_2467delTG (p.C822X) and in MLH1: c.1023delG (p.R341Xfs), c.1127_1128dupAT (p.K377Xfs) and c.1310delC (p.P437Xfs); three mutations leading to splice aberrations, in MSH2: c.1661G>C (r.1511_1661del) and in MLH1: c.677+3A>C (r.589_677del) and c.1990-2A>G predicted to result in a splice site defect. The remaining two mutations are unclassified variants with assumed pathogenicity: one missense mutation in the highly conserved ATPase domain of MLH1 (c.122A>G [p.D41G]) and one in-frame insertion of twelve nucleotides in MLH1 (c.2155_2156insATGTGTTCCACA [p.I719delinsNVFHI]). These two mutations were not found in 102 alleles of healthy control individuals. The corresponding tumors from all patients showed a high level of microsatellite instability (MSI-H). Immunohistochemistry (IHC) revealed complete loss of expression of the affected protein in the tumor cells from all but three patients. The tumors from the patients with the mutations c.1127_1128dupAT and c.1990-2A>G showed a reduction of expression of the MLH1-protein, rather than complete loss. In the tumor from the patient with the missense mutation c.122A>G [p.D41G] a normal expression of the proteins coded by MLH1 and MSH2 was noticed.


Assuntos
Neoplasias Colorretais Hereditárias sem Polipose/genética , Proteínas de Ligação a DNA/genética , Mutação em Linhagem Germinativa , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Alelos , Pareamento Incorreto de Bases , Proteínas de Transporte , Códon sem Sentido , Neoplasias Colorretais Hereditárias sem Polipose/epidemiologia , Feminino , Mutação da Fase de Leitura , Alemanha/epidemiologia , Humanos , Masculino , Repetições de Microssatélites , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Proteína 2 Homóloga a MutS , Mutagênese Insercional , Proteínas Nucleares , Sítios de Splice de RNA/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA