Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 113: 66-82, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37369341

RESUMO

Stress-induced ß2-adrenergic receptor (ß2AR) activation in B cells increases IgG secretion; however, the impact of this activation on antibody affinity and the underlying mechanisms remains unclear. In the current study, we demonstrate that stress in mice following ovalbumin (OVA) or SARS-CoV-2 RBD immunization significantly increases both serum and surface-expressed IgG binding to the immunogen, while concurrently reducing surface IgG expression and B cell clonal expansion. These effects were abolished by pharmacological ß2AR blocking or when the experiments were conducted in ß2AR -/- mice. In the second part of our study, we used single B cell sorting to characterize the monoclonal antibodies (mAbs) generated following ß2AR activation in cultured RBD-stimulated B cells from convalescent SARS-CoV-2 donors. Ex vivo ß2AR activation increased the affinities of the produced anti-RBD mAbs by 100-fold compared to mAbs produced by the same donor control cultures. Consistent with the mouse experiments, ß2AR activation reduced both surface IgG levels and the frequency of expanded clones. mRNA sequencing revealed a ß2AR-dependent upregulation of the PI3K pathway and B cell receptor (BCR) signaling through AKT phosphorylation, as well as an increased B cell motility. Overall, our study demonstrates that stress-mediated ß2AR activation drives changes in B cells associated with BCR activation and higher affinity antibodies.


Assuntos
Adrenérgicos , COVID-19 , Camundongos , Animais , Fosfatidilinositol 3-Quinases , SARS-CoV-2/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Imunoglobulina G
2.
Nat Commun ; 12(1): 602, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33504803

RESUMO

Mycobacterium tuberculosis (Mtb) exposure drives antibody responses, but whether patients with active tuberculosis elicit protective antibodies, and against which antigens, is still unclear. Here we generate monoclonal antibodies from memory B cells of one patient to investigate the B cell responses during active infection. The antibodies, members of four distinct B cell clones, are directed against the Mtb phosphate transporter subunit PstS1. Antibodies p4-36 and p4-163 reduce Mycobacterium bovis-BCG and Mtb levels in an ex vivo human whole blood growth inhibition assay in an FcR-dependent manner; meanwhile, germline versions of p4-36 and p4-163 do not bind Mtb. Crystal structures of p4-36 and p4-170, complexed to PstS1, are determined at 2.1 Å and 2.4 Å resolution, respectively, to reveal two distinctive PstS1 epitopes. Lastly, a prophylactic p4-36 and p4-163 treatment in Mtb-infected Balb/c mice reduces bacterial lung burden by 50%. Our study shows that inhibitory anti-PstS1 B cell responses arise during active tuberculosis.


Assuntos
Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Membrana Transportadoras/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Tuberculose/prevenção & controle , Adulto , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Linfócitos B/imunologia , Proteínas de Bactérias/química , Epitopos/química , Humanos , Memória Imunológica , Masculino , Camundongos Endogâmicos BALB C , Modelos Moleculares , Estrutura Secundária de Proteína , Relação Estrutura-Atividade , Células THP-1 , Tuberculose/sangue , Tuberculose/microbiologia
3.
Int J Cancer ; 145(9): 2521-2534, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31216364

RESUMO

The major cause of melanoma mortality is metastasis to distant organs, including lungs and brain. Reciprocal interactions of metastasizing tumor cells with stromal cells in secondary sites play a critical role in all stages of tumorigenesis and metastasis. Changes in the metastatic microenvironment were shown to precede clinically relevant metastases, and may occur prior to the arrival of disseminated tumor cells to the distant organ, thus creating a hospitable "premetastatic niche." Exosomes secreted by tumor cells were demonstrated to play an important role in the preparation of a hospitable metastatic niche. However, the functional role of melanoma-derived exosomes on metastatic niche formation, and the downstream pathways activated in stromal cells at the metastatic niche are largely unresolved. Here we show that extracellular vesicles (EVs) secreted by metastatic melanoma cells that spontaneously metastasize to lungs and to brain, activate proinflammatory signaling in lung fibroblasts and in astrocytes. Interestingly, unlike paracrine signaling by melanoma cells, EVs secreted by metastatic melanoma cells instigated a proinflammatory gene signature in lung fibroblasts but did not activate wound-healing functions, suggesting that tumor cell-secreted EVs activate distinct CAF characteristics and tumor-promoting functions. Moreover, melanoma-secreted EVs also activated proinflammatory signaling in astrocytes, indicating that EV-mediated reprogramming of stromal cells is a general mechanism of modulating the metastatic niche in multiple distant organs. Thus, our study demonstrates that melanoma-derived EVs reprogram tumor-promoting functions in stromal cells in a distinct manner, implicating a central role for tumor-derived EV signaling in promoting the formation of an inflammatory metastatic niche.


Assuntos
Vesículas Extracelulares/patologia , Inflamação/patologia , Melanoma/patologia , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Animais , Astrócitos/patologia , Exossomos/patologia , Fibroblastos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Comunicação Parácrina/fisiologia , Células Estromais/patologia
4.
J Exp Med ; 215(12): 3075-3093, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30470719

RESUMO

Cancer-associated fibroblasts (CAFs) are highly prominent in breast tumors, but their functional heterogeneity and origin are still largely unresolved. We report that bone marrow (BM)-derived mesenchymal stromal cells (MSCs) are recruited to primary breast tumors and to lung metastases and differentiate to a distinct subpopulation of CAFs. We show that BM-derived CAFs are functionally important for tumor growth and enhance angiogenesis via up-regulation of Clusterin. Using newly generated transgenic mice and adoptive BM transplantations, we demonstrate that BM-derived fibroblasts are a substantial source of CAFs in the tumor microenvironment. Unlike resident CAFs, BM-derived CAFs do not express PDGFRα, and their recruitment resulted in a decrease in the percentage of PDGFRα-expressing CAFs. Strikingly, decrease in PDGFRα in breast cancer patients was associated with worse prognosis, suggesting that BM-derived CAFs may have deleterious effects on survival. Therefore, PDGFRα expression distinguishes two functionally unique CAF populations in breast tumors and metastases and may have important implications for patient stratification and precision therapeutics.


Assuntos
Células da Medula Óssea/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias Mamárias Animais/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas de Neoplasias/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Microambiente Tumoral , Animais , Células da Medula Óssea/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Fibroblastos , Humanos , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Proteínas de Neoplasias/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
5.
Cancer Res ; 76(15): 4359-71, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27261506

RESUMO

Malignant melanoma is the deadliest of skin cancers. Melanoma frequently metastasizes to the brain, resulting in dismal survival. Nevertheless, mechanisms that govern early metastatic growth and the interactions of disseminated metastatic cells with the brain microenvironment are largely unknown. To study the hallmarks of brain metastatic niche formation, we established a transplantable model of spontaneous melanoma brain metastasis in immunocompetent mice and developed molecular tools for quantitative detection of brain micrometastases. Here we demonstrate that micrometastases are associated with instigation of astrogliosis, neuroinflammation, and hyperpermeability of the blood-brain barrier. Furthermore, we show a functional role for astrocytes in facilitating initial growth of melanoma cells. Our findings suggest that astrogliosis, physiologically instigated as a brain tissue damage response, is hijacked by tumor cells to support metastatic growth. Studying spontaneous melanoma brain metastasis in a clinically relevant setting is the key to developing therapeutic approaches that may prevent brain metastatic relapse. Cancer Res; 76(15); 4359-71. ©2016 AACR.


Assuntos
Astrócitos/patologia , Melanoma/complicações , Animais , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Humanos , Inflamação , Melanoma/patologia , Camundongos , Metástase Neoplásica , Neovascularização Patológica/patologia , Neoplasias Cutâneas , Melanoma Maligno Cutâneo
6.
ACS Nano ; 8(3): 1958-65, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24660817

RESUMO

The treatment of neurodegenerative diseases remains a tremendous challenge due to the limited access of molecules across the blood-brain barrier, especially large molecules such as peptides and proteins. As a result, at most, a small percentage of a drug that is administered systemically will reach the central nervous system in its active form. Currently, research in the field focuses on developing safer and more effective approaches to deliver peptides and proteins into the central nervous system. Multiple strategies have been developed for this purpose. However, noninvasive approaches, such as nanostructured protein delivery carriers and intranasal administration, seem to be the most promising strategies for the treatment of chronic diseases, which require long-term interventions. These approaches are both target-specific and able to rapidly bypass the blood-brain barrier. In this Perspective, we detail some of these strategies and discuss some of the potential pitfalls and opportunities in this field. The next generation strategies will most likely be more cell-type-specific. Devising these strategies to target the brain may ultimately become a novel therapeutic modality to treat neurodegenerative diseases.


Assuntos
Nanomedicina/métodos , Doenças Neurodegenerativas/terapia , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Humanos
7.
Clin Cancer Res ; 17(13): 4254-66, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21571866

RESUMO

PURPOSE: Klotho is a transmembrane protein which can be shed, act as a circulating hormone and modulate the insulin-like growth factor (IGF)-I and the fibroblast growth factor (FGF) pathways. We have recently identified klotho as a tumor suppressor in breast cancer. Klotho is expressed in the normal pancreas and both the IGF-I and FGF pathways are involved in pancreatic cancer development. We, therefore, undertook to study the expression and activity of klotho in pancreatic cancer. EXPERIMENTAL DESIGN: Klotho expression was studied using immunohistochemistry and quantitative RT-PCR. Effects of klotho on cell growth were assessed in the pancreatic cancer cells Panc1, MiaPaCa2, and Colo357, using colony and MTT assays and xenograft models. Signaling pathway activity was measured by Western blotting. RESULTS: Klotho expression is downregulated in pancreatic adenocarcinoma. Overexpression of klotho, or treatment with soluble klotho, reduced growth of pancreatic cancer cells in vitro and in vivo, and inhibited activation of the IGF-I and the bFGF pathways. KL1 is a klotho subdomain formed by cleavage or alternative splicing. Compared with the full-length protein, KL1 showed similar growth inhibitory activity but did not promote FGF23 signaling. Thus, its administration to mice showed favorable safety profile. CONCLUSIONS: These studies indicate klotho as a potential tumor suppressor in pancreatic cancer, and suggest, for the first time, that klotho tumor suppressive activities are mediated through its KL1 domain. These results suggest the use of klotho or KL1 as potential strategy for the development of novel therapeutic interventions for pancreatic cancer.


Assuntos
Adenocarcinoma , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Glucuronidase/metabolismo , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Neoplasias Pancreáticas , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Regulação para Baixo/genética , Feminino , Fator 2 de Crescimento de Fibroblastos/genética , Fator de Crescimento de Fibroblastos 23 , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glucuronidase/genética , Células HEK293 , Humanos , Fator de Crescimento Insulin-Like I/genética , Proteínas Klotho , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Supressoras de Tumor/genética
8.
Cancer Res ; 68(17): 6978-86, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18757412

RESUMO

Studies have conjectured that nonsteroidal anti-inflammatory drugs (NSAID) inhibit growth of various malignancies by inhibiting cyclooxygenase-2 (COX-2) enzyme activity. Yet, several lines of evidence indicate that a COX-2-independent mechanism may also be involved in their antitumor effects. Here, we report that NSAIDs may inhibit the growth of glioblastoma multiforme (GBM) cells through COX-2-independent mechanisms, including up-regulation of both 15-hydroxyprostaglandin dehydrogenase (15-PGDH, the key prostaglandin catabolic enzyme) and the cell cycle inhibitor p21. Using Western blot and real-time PCR analysis in various GBM cell lines, we observed up-regulation of 15-PGDH and p21 after NSAIDs treatment. To elucidate the role of 15-PGDH in GBM, transfection assays were conducted using the T98G GBM cell line. Overexpression of 15-PGDH suppressed cell growth and was associated with increased expression of p21. In an attempt to investigate the roles of COX-2, 15-PGDH, and p21 in the inhibition of growth of GBM, small interfering RNA (siRNA) against each of these proteins was transfected into T98G cells. Inhibition of growth mediated by NSAIDs was partially reversed after knockdown of either 15-PGDH or p21, but not after COX-2 knockdown. Moreover, expression level of p21 was not affected in COX-2 siRNA transfected cells. Our studies provide evidence that the up-regulation of 15-PGDH induced by NSAIDs has the potential to inhibit growth of GBM, in part, by up-regulation of p21 possibly independent from COX-2 enzymatic function.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias Encefálicas/patologia , Divisão Celular/efeitos dos fármacos , Glioma/patologia , Hidroxiprostaglandina Desidrogenases/metabolismo , Sequência de Bases , Neoplasias Encefálicas/enzimologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Primers do DNA , Regulação para Baixo , Glioma/enzimologia , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
9.
J Biol Chem ; 283(1): 110-119, 2008 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17984088

RESUMO

Restriction of glutamine synthetase to the nervous system is mainly achieved through the mutual function of the glucocorticoid receptor and the neural restrictive silencing factor, NRSF/REST. Glucocorticoids induce glutamine synthetase expression in neural tissues while NRSF/REST represses the hormonal response in non-neural cells. NRSF/REST is a modular protein that contains two independent repression domains, at the N and C termini of the molecule, and is dominantly expressed in nonneural cells. Neural tissues express however splice variants, REST4/5, which contain the repression domain at the N, but not at the C terminus of the molecule. Here we show that full-length NRSF/REST or its C-terminal domain can inhibit almost completely the induction of gene transcription by glucocorticoids. By contrast, the N-terminal domain not only fails to repress the hormonal response but rather stimulates it markedly. The inductive activity of the N-terminal domain is mediated by hBrm, which is recruited to the promoter only in the concomitant presence of GR. Importantly, a similar inductive activity is also exerted by the splice variant REST4. These findings raise the possibility that NRSF/REST exhibits a dual role in regulation of glutamine synthetase. It represses gene induction in nonneural cells and enhances the hormonal response, via its splice variant, in the nervous system.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Proteínas Repressoras/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sítios de Ligação/genética , Western Blotting , Células COS , Linhagem Celular Tumoral , Células Cultivadas , Galinhas , Chlorocebus aethiops , Vetores Genéticos/genética , Células HeLa , Humanos , Imunoprecipitação , Regiões Promotoras Genéticas/genética , Receptores de Glucocorticoides/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retina/citologia , Retina/efeitos dos fármacos , Retina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional , Leveduras/genética , Leveduras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA