Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Blood ; 140(26): 2805-2817, 2022 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-36283106

RESUMO

Myelofibrosis (MF) is a disease associated with high unmet medical needs because allogeneic stem cell transplantation is not an option for most patients, and JAK inhibitors are generally effective for only 2 to 3 years and do not delay disease progression. MF is characterized by dysplastic megakaryocytic hyperplasia and progression to fulminant disease, which is associated with progressively increasing marrow fibrosis. Despite evidence that the inflammatory milieu in MF contributes to disease progression, the specific factors that promote megakaryocyte growth are poorly understood. Here, we analyzed changes in the cytokine profiles of MF mouse models before and after the development of fibrosis, coupled with the analysis of bone marrow populations using single-cell RNA sequencing. We found high interleukin 13 (IL-13) levels in the bone marrow of MF mice. IL-13 promoted the growth of mutant megakaryocytes and induced surface expression of transforming growth factor ß and collagen biosynthesis. Similarly, analysis of samples from patients with MF revealed elevated levels of IL-13 in the plasma and increased IL-13 receptor expression in marrow megakaryocytes. In vivo, IL-13 overexpression promoted disease progression, whereas reducing IL-13/IL-4 signaling reduced several features of the disease, including fibrosis. Finally, we observed an increase in the number of marrow T cells and mast cells, which are known sources of IL-13. Together, our data demonstrate that IL-13 is involved in disease progression in MF and that inhibition of the IL-13/IL-4 signaling pathway might serve as a novel therapeutic target to treat MF.


Assuntos
Transtornos Mieloproliferativos , Neoplasias , Mielofibrose Primária , Camundongos , Animais , Interleucina-13/uso terapêutico , Interleucina-4 , Neoplasias/complicações , Transtornos Mieloproliferativos/complicações , Mielofibrose Primária/genética , Transdução de Sinais/genética , Fibrose , Progressão da Doença
3.
Biomolecules ; 12(2)2022 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-35204735

RESUMO

Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-ß1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.


Assuntos
Citocinas , Fator de Transcrição GATA1 , Mielofibrose Primária , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Citocinas/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fator de Transcrição GATA1/metabolismo , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Mielofibrose Primária/metabolismo , Mielofibrose Primária/patologia
4.
Hematol Oncol Clin North Am ; 35(2): 191-203, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33641863

RESUMO

Megakaryocytes give rise to platelets, which have a wide variety of functions in coagulation, immune response, inflammation, and tissue repair. Dysregulation of megakaryocytes is a key feature of in the myeloproliferative neoplasms, especially myelofibrosis. Megakaryocytes are among the main drivers of myelofibrosis by promoting myeloproliferation and bone marrow fibrosis. In vivo targeting of megakaryocytes by genetic and pharmacologic approaches ameliorates the disease, underscoring the important role of megakaryocytes in myeloproliferative neoplasms. Here we review the current knowledge of the function of megakaryocytes in the JAK2, CALR, and MPL-mutant myeloproliferative neoplasms.


Assuntos
Megacariócitos , Transtornos Mieloproliferativos , Neoplasias , Mielofibrose Primária , Calreticulina/genética , Humanos , Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/genética , Neoplasias/genética , Mielofibrose Primária/genética , Receptores de Trombopoetina/genética
5.
JCI Insight ; 4(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30843874

RESUMO

Treg differentiation, maintenance, and function are controlled by the transcription factor FoxP3, which can be destabilized under inflammatory or other pathological conditions. Tregs can be destabilized under inflammatory or other pathological conditions, but the underlying mechanisms are not fully defined. Herein, we show that inflammatory cytokines induce ER stress response, which destabilizes Tregs by suppressing FoxP3 expression, suggesting a critical role of the ER stress response in maintaining Treg stability. Indeed, genetic deletion of Hrd1, an E3 ligase critical in suppressing the ER stress response, leads to elevated expression of ER stress-responsive genes in Treg and largely diminishes Treg suppressive functions under inflammatory condition. Mice with Treg-specific ablation of Hrd1 displayed massive multiorgan lymphocyte infiltration, body weight loss, and the development of severe small intestine inflammation with aging. At the molecular level, the deletion of Hrd1 led to the activation of both the ER stress sensor IRE1α and its downstream MAPK p38. Pharmacological suppression of IRE1α kinase, but not its endoribonuclease activity, diminished the elevated p38 activation and fully rescued the stability of Hrd1-null Tregs. Taken together, our studies reveal ER stress response as a previously unappreciated mechanism underlying Treg instability and that Hrd1 is crucial for maintaining Treg stability and functions through suppressing the IRE1α-mediated ER stress response.


Assuntos
Citocinas/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose , Colite/imunologia , Colite/patologia , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Endorribonucleases , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Homeostase , Inflamação/imunologia , Inflamação/patologia , Linfócitos Nulos , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Transcriptoma , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética
6.
EMBO J ; 37(22)2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30389664

RESUMO

The endoplasmic reticulum-associated protein degradation (ERAD) is responsible for recognizing and retro-translocating protein substrates, misfolded or not, from the ER for cytosolic proteasomal degradation. HMG-CoA Reductase (HMGCR) Degradation protein-HRD1-was initially identified as an E3 ligase critical for ERAD. However, its physiological functions remain largely undefined. Herein, we discovered that hepatic HRD1 expression is induced in the postprandial condition upon mouse refeeding. Mice with liver-specific HRD1 deletion failed to repress FGF21 production in serum and liver even in the refeeding condition and phenocopy the FGF21 gain-of-function mice showing growth retardation, female infertility, and diurnal circadian behavior disruption. HRD1-ERAD facilitates the degradation of the liver-specific ER-tethered transcription factor CREBH to downregulate FGF21 expression. HRD1-ERAD catalyzes polyubiquitin conjugation onto CREBH at lysine 294 for its proteasomal degradation, bridging a multi-organ crosstalk in regulating growth, circadian behavior, and female fertility through regulating the CREBH-FGF21 regulatory axis.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Degradação Associada com o Retículo Endoplasmático , Fatores de Crescimento de Fibroblastos/biossíntese , Fígado/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Feminino , Fertilidade/genética , Fatores de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Poliubiquitina/genética , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitina-Proteína Ligases/genética
7.
Nat Commun ; 9(1): 3659, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30201971

RESUMO

The HMG-CoA reductase degradation protein 1 (HRD1) has been identified as a key enzyme for endoplasmic reticulum-associated degradation of misfolded proteins, but its organ-specific physiological functions remain largely undefined. Here we show that mice with HRD1 deletion specifically in the liver display increased energy expenditure and are resistant to HFD-induced obesity and liver steatosis and insulin resistance. Proteomic analysis identifies a HRD1 interactome, a large portion of which includes metabolic regulators. Loss of HRD1 results in elevated ENTPD5, CPT2, RMND1, and HSD17B4 protein levels and a consequent hyperactivation of both AMPK and AKT pathways. Genome-wide mRNA sequencing revealed that HRD1-deficiency reprograms liver metabolic gene expression profiles, including suppressing genes involved in glycogenesis and lipogenesis and upregulating genes involved in glycolysis and fatty acid oxidation. We propose HRD1 as a liver metabolic regulator and a potential drug target for obesity, fatty liver disease, and insulin resistance associated with the metabolic syndrome.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Fígado/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adenilato Quinase/metabolismo , Animais , Peso Corporal , Dieta Hiperlipídica , Ativação Enzimática , Ácidos Graxos/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Glicólise , Células HEK293 , Células Hep G2 , Humanos , Lipogênese , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Proteoma , Proteômica , Triglicerídeos/metabolismo , Ubiquitinação
8.
J Biol Chem ; 293(33): 12934-12944, 2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-29907570

RESUMO

Humoral immunity involves multiple checkpoints that occur in B cell development, maturation, and activation. The pre-B-cell receptor (pre-BCR) is expressed following the productive recombination of the immunoglobulin heavy-chain gene, and sSignalsing through the pre-BCR are required for the differentiation of pre-B cells into immature B cells. However, the molecular mechanisms controlling the pre-BCR expression and signaling strength remain undefined. Herein, we probed the role of the endoplasmic reticulum-associated, stress-activated E3 ubiquitin ligase HMG-CoA reductase degradation 1 (Hrd1) in B cell differentiation. Using mice with a specific Hrd1 deletion in pro-B cells and subsequent B cell developmental stages, we showed that the E3 ubiquitin ligase Hrd1 governs a critical checkpoint during B cell development. We observed that Hrd1 is required for degradation of the pre-BCR complex during the early stage of B cell development. As a consequence, loss of Hrd1 in the B cell lineage resulted in increased pre-BCR expression levels and a developmental defect in the transition from large to small pre-B cells. This defect, in turn, resulted in reduced fewer mature B cells in bone marrow and peripheral lymphoid organs. Our results revealed a novel critical role of Hrd1 in controlling a critical checkpoint in B cell-mediated immunity and suggest that Hrd1 may functioning as an E3 ubiquitin ligase of the pre-BCR complex.


Assuntos
Células da Medula Óssea/imunologia , Diferenciação Celular/imunologia , Retículo Endoplasmático/imunologia , Células Precursoras de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/enzimologia , Diferenciação Celular/genética , Linhagem Celular , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/enzimologia , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
9.
Blood ; 132(4): 423-434, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29844011

RESUMO

Ras mutations are commonly observed in juvenile myelomonocytic leukemia (JMML) and chronic myelomonocytic leukemia (CMML). JMML and CMML transform into acute myeloid leukemia (AML) in about 10% and 50% of patients, respectively. However, how additional events cooperate with Ras to promote this transformation are largely unknown. We show that absence of the ubiquitin-specific peptidase 22 (USP22), a component of the Spt-Ada-GCN5-acetyltransferase chromatin-remodeling complex that is linked to cancer progression, unexpectedly promotes AML transformation in mice expressing oncogenic KrasG12D/+ USP22 deficiency in KrasG12D/+ mice resulted in shorter survival compared with control mice. This was due to a block in myeloid cell differentiation leading to the generation of AML. This effect was cell autonomous because mice transplanted with USP22-deficient KrasG12D/+ cells developed an aggressive disease and died rapidly. The transcriptome profile of USP22-deficient KrasG12D/+ progenitors resembled leukemic stem cells and was highly correlated with genes associated with poor prognosis in AML. We show that USP22 functions as a PU.1 deubiquitylase by positively regulating its protein stability and promoting the expression of PU.1 target genes. Reconstitution of PU.1 overexpression in USP22-deficient KrasG12D/+ progenitors rescued their differentiation. Our findings uncovered an unexpected role for USP22 in Ras-induced leukemogenesis and provide further insights into the function of USP22 in carcinogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Endopeptidases/fisiologia , Leucemia Mieloide/patologia , Leucemia Mielomonocítica Juvenil/patologia , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/metabolismo , Animais , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Perfilação da Expressão Gênica , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Prognóstico , Proteínas Proto-Oncogênicas/genética , Taxa de Sobrevida , Transativadores/genética , Ubiquitina Tiolesterase
10.
Proc Natl Acad Sci U S A ; 113(37): 10394-9, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27573825

RESUMO

Humoral immunity involves multiple checkpoints during B-cell development, maturation, and activation. The cell death receptor CD95/Fas-mediated apoptosis plays a critical role in eliminating the unwanted activation of B cells by self-reactive antigens and in maintaining B-cell homeostasis through activation-induced B-cell death (AICD). The molecular mechanisms controlling AICD remain largely undefined. Herein, we show that the E3 ubiquitin ligase Hrd1 protected B cells from activation-induced cell death by degrading the death receptor Fas. Hrd1-null B cells exhibited high Fas expression during activation and rapidly underwent Fas-mediated apoptosis, which could be largely inhibited by FasL neutralization. Fas mutation in Hrd1 KO mice abrogated the increase in B-cell AICD. We identified Hrd1 as the first E3 ubiquitin ligase of the death receptor Fas and Hrd1-mediated Fas destruction as a molecular mechanism in regulating B-cell immunity.


Assuntos
Proteína Ligante Fas/genética , Ativação Linfocitária/genética , Ubiquitina-Proteína Ligases/genética , Receptor fas/genética , Animais , Apoptose/genética , Apoptose/imunologia , Linfócitos B/imunologia , Retículo Endoplasmático/enzimologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Proteólise , Ubiquitina-Proteína Ligases/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Receptor fas/metabolismo
11.
Nat Commun ; 7: 12073, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27417417

RESUMO

Identification of positive regulators of T-cell immunity induced during autoimmune diseases is critical for developing novel therapies. The endoplasmic reticulum resident ubiquitin ligase Hrd1 has recently emerged as a critical regulator of dendritic cell antigen presentation, but its role in T-cell immunity is unknown. Here we show that genetic deletion of Hrd1 in mice inhibits T-cell proliferation, production of IL-2, and differentiation of Th1 and Th17 cells, and consequently protects mice from experimental autoimmune encephalomyelitis. Hrd1 facilitates T-cell proliferation by the destruction of cyclin-dependent kinase inhibitor p27(kip1), and deletion of p27(kip1) in Hrd1-null T-cells rescues proliferative capacity but not the production of cytokines, including IL-2, IFN-γ and IL-17. T-cell expression of Hrd1 is higher in patients with multiple sclerosis than in healthy individuals, and knockdown of Hrd1 in human CD4(+) T cells inhibits activation and differentiation to Th1 and Th17 cells. Our study identifies Hrd1 as a previously unappreciated positive regulator of T cells and implies that Hrd1 is a potential therapeutic target for autoimmune diseases.


Assuntos
Retículo Endoplasmático/metabolismo , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linfócitos T CD4-Positivos/imunologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Humanos , Membranas Intracelulares/metabolismo , Ativação Linfocitária/imunologia , Camundongos Knockout , Camundongos Transgênicos , Esclerose Múltipla/imunologia , Linfócitos T/metabolismo , Células Th1/imunologia , Células Th17/imunologia , Ubiquitina-Proteína Ligases/genética
12.
Oncotarget ; 7(28): 44848-44856, 2016 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-27057639

RESUMO

Deubiquitylases remove ubiquitin moieties from different substrates to regulate protein activity and cell homeostasis. Since this posttranslational modification plays a role in several different cellular functions, its deregulation has been associated with different pathologies. Aberrant expression of the Ubiquitin-Specific Peptidase 22 (USP22) has been associated with poor cancer prognosis and neurological disorders. However, little is known about USP22 role in these pathologies or in normal physiology. This review summarizes the current knowledge about USP22 function from yeast to human and provides an overview of the possible mechanisms by which USP22 is emerging as a potential oncogene.


Assuntos
Ciclo Celular/genética , Neoplasias/genética , Oncogenes/genética , Tioléster Hidrolases/genética , Antineoplásicos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tioléster Hidrolases/antagonistas & inibidores , Tioléster Hidrolases/metabolismo , Ubiquitina Tiolesterase
13.
Oncotarget ; 7(3): 2809-22, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26646452

RESUMO

The CXCR4 receptor (Chemokine C-X-C motif receptor 4) is highly expressed in different hematological malignancies including chronic lymphocytic leukemia (CLL). The CXCR4 ligand (CXCL12) stimulates CXCR4 promoting cell survival and proliferation, and may contribute to the tropism of leukemia cells towards lymphoid tissues. Therefore, strategies targeting CXCR4 may constitute an effective therapeutic approach for CLL. To address that question, we studied the effect of Ulocuplumab (BMS-936564), a fully human IgG4 anti-CXCR4 antibody, using a stroma--CLL cells co-culture model. We found that Ulocuplumab (BMS-936564) inhibited CXCL12 mediated CXCR4 activation-migration of CLL cells at nanomolar concentrations. This effect was comparable to AMD3100 (Plerixafor--Mozobil), a small molecule CXCR4 inhibitor. However, Ulocuplumab (BMS-936564) but not AMD3100 induced apoptosis in CLL at nanomolar concentrations in the presence or absence of stromal cell support. This pro-apoptotic effect was independent of CLL high-risk prognostic markers, was associated with production of reactive oxygen species and did not require caspase activation. Overall, these findings are evidence that Ulocuplumab (BMS-936564) has biological activity in CLL, highlight the relevance of the CXCR4-CXCL12 pathway as a therapeutic target in CLL, and provide biological rationale for ongoing clinical trials in CLL and other hematological malignancies.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Quimiocina CXCL12/biossíntese , Imino Furanoses/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Pirimidinonas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores CXCR4/antagonistas & inibidores , Actinas/metabolismo , Benzilaminas , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular , Quimiocina CXCL12/metabolismo , Ciclamos , Ativação Enzimática/efeitos dos fármacos , Compostos Heterocíclicos/farmacologia , Humanos , Células Jurkat , Leucócitos Mononucleares , Receptores CXCR4/biossíntese , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
15.
Hum Gene Ther ; 26(1): 14-25, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25382101

RESUMO

Ad-ISF35, an adenovirus vector encoding a membrane-bound engineered CD154 chimeric protein (ISF35), induces complete A20 lymphoma tumor regression in mice after intratumoral direct injection (IDI). Ad-ISF35 induced durable local and systemic antitumor responses associated with a rapid tumor infiltration of macrophages and neutrophils as well as increased levels of proinflammatory cytokines in the tumor microenvironment. Ad-ISF35 IDI transduced preferentially fibroblasts and macrophages present in the tumor microenvironment, and ISF35 protein expression was observed in only 0.25% of cells present in the tumor. Moreover, Ad-ISF35 IDI induced upregulation of CD40 in tumor and immune regulatory cells, including those that did not express ISF35, suggesting the presence of a strong bystander effect. These responses resulted in the generation of IFN-γ-secreting cytotoxic lymphocytes and the production of specific cytotoxic antibodies against lymphoma cells. Overall, cellular immune therapy based on ISF35 induced phenotypic changes in the tumor cells and tumor microenvironment that were associated with a break in tumor immune tolerance and a curative antitumor effect in this lymphoma mouse model. Our data highlight the potential activity that modulation of costimulatory signaling has in cancer therapy.


Assuntos
Adenoviridae/genética , Ligante de CD40/genética , Vetores Genéticos/genética , Tolerância Imunológica , Linfoma/genética , Linfoma/imunologia , Proteínas Recombinantes de Fusão/genética , Animais , Ligante de CD40/imunologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Imunidade Humoral , Mediadores da Inflamação/metabolismo , Injeções Intralesionais , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfoma/mortalidade , Linfoma/patologia , Linfoma/terapia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Necrose/imunologia , Necrose/patologia , Infiltração de Neutrófilos , Proteínas Recombinantes de Fusão/imunologia , Transgenes , Carga Tumoral , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
16.
J Cell Sci ; 127(Pt 12): 2683-96, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24741066

RESUMO

Nuclear lamins form the major structural elements that comprise the nuclear lamina. Loss of nuclear structural integrity has been implicated as a key factor in the lamin A/C gene mutations that cause laminopathies, whereas the normal regulation of lamin A assembly and organization in interphase cells is still undefined. We assumed phosphorylation to be a major determinant, identifying 20 prime interphase phosphorylation sites, of which eight were high-turnover sites. We examined the roles of these latter sites by site-directed mutagenesis, followed by detailed microscopic analysis - including fluorescence recovery after photobleaching, fluorescence correlation spectroscopy and nuclear extraction techniques. The results reveal three phosphorylation regions, each with dominant sites, together controlling lamin A structure and dynamics. Interestingly, two of these interphase sites are hyper-phosphorylated in mitotic cells and one of these sites is within the sequence that is missing in progerin of the Hutchinson-Gilford progeria syndrome. We present a model where different phosphorylation combinations yield markedly different effects on the assembly, subunit turnover and the mobility of lamin A between, and within, the lamina, the nucleoplasm and the cytoplasm of interphase cells.


Assuntos
Interfase , Lamina Tipo A/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Células HeLa , Humanos , Dados de Sequência Molecular , Lâmina Nuclear/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Transporte Proteico , Fatores de Transcrição
17.
Cancer Res ; 72(12): 2937-48, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22505652

RESUMO

New therapies for chronic lymphocytic leukemia (CLL) are needed, particularly those that can eradicate residual disease and elicit anti-CLL immune responses. CD40 ligation on CLL cells, which can be achieved using adenovirus encoding chimeric CD154 (Ad-ISF35), enhances their ability to function as antigen-presenting cells and increases their sensitivity to clearance by immune-effector mechanisms. In this study, we report the results of a first-in-man phase I trial of intranodal direct injection (IDI) of Ad-ISF35 in patients with CLL to evaluate toxicity, safety, and tolerability. Fifteen patients received a single IDI of 1 × 10(10) to 33 × 10(10) Ad-ISF35 viral particles (vp), with a defined maximum tolerated dose as 1 × 10(11) vp. Although the most common adverse events were transient grade 1 to 2 pain at the injection site and flu-like symptoms following IDI, some patients receiving the highest dose had transient, asymptomatic grade 3 to 4 hypophosphatemia, neutropenia, or transaminitis. Increased expression of death receptor, immune costimulatory molecules, and Ad-ISF35 vector DNA was detected in circulating CLL cells. Notably, we also observed preliminary clinical responses, including reductions in leukemia cell counts, lymphadenopathy, and splenomegaly. Six patients did not require additional therapy for more than 6 months, and three achieved a partial remission. In conclusion, Ad-ISF35 IDI was safely delivered in patients with CLLs and induced systemic biologic and clinical responses. These results provide the rationale for phase II studies in CLLs, lymphomas, and CD40-expressing solid tumors.


Assuntos
Ligante de CD40/genética , Ligante de CD40/imunologia , Terapia Genética , Leucemia Linfocítica Crônica de Células B/terapia , Adenoviridae/genética , Idoso , Formação de Anticorpos , Células Apresentadoras de Antígenos/imunologia , Citocinas/sangue , Feminino , Vetores Genéticos , Humanos , Imunoterapia , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/imunologia , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/imunologia
18.
Amino Acids ; 42(6): 2165-75, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21674161

RESUMO

Peptide 11389 from CD21-binding region of EBV-gp350/220 protein binds to PBMCs inducing IL-6 expression and inhibiting EBV-binding to PBMCs. In addition, anti-peptide 11389 antibodies recognize EBV-infected cells and inhibit both EBV infection and IL-6 production in PBMCs. We have postulated that native structure stabilization of peptide 11389 sequence can increase its biological activity. The strategy was to modify its sequence to restrict the number of structures that peptide 11389 could acquire in solution (decreasing peptide's configurational entropy) and to weaken the non-relevant intermolecular interactions (decreasing its hydrophobicity), preserving CD21-interacting residues and structure as displayed in the native protein. Thirteen analog peptides were designed and synthesized; most of them were monomers containing an intra-chain disulfide bridge. Analog peptides 34058, 34060, 34061, 34296, 34298, 34299 and 34300 inhibited EBV invasion of PBMCs. Peptides 34059, 34060, 34295 and 34297 induced IL-6 levels in PBMCs (EC50=3.4, 3.3, 0.5, 0.5 µM, respectively) at higher potency than peptide 11389 (EC50=5.8 µM). Peptides 34057, 34059, 34060, 34301 and 34302 interacted with anti-EBV antibodies with affinities from 3 to 50 times higher than peptide 11389. Most of analog peptides were highly immunogenic and elicited antibodies that cross-react with EBV. In conclusion, we have designed peptides displaying higher biological activity than peptide 11389.


Assuntos
Antígenos Virais/imunologia , Herpesvirus Humano 4/imunologia , Leucócitos Mononucleares/imunologia , Peptídeos/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , Antígenos Virais/química , Antígenos Virais/genética , Relação Dose-Resposta a Droga , Entropia , Herpesvirus Humano 4/química , Herpesvirus Humano 4/genética , Humanos , Interações Hidrofóbicas e Hidrofílicas , Interleucina-6/biossíntese , Interleucina-6/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/farmacologia , Conformação Proteica , Receptores de Complemento 3d/imunologia , Receptores de Complemento 3d/metabolismo , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética , Internalização do Vírus/efeitos dos fármacos
19.
Amino Acids ; 39(5): 1507-19, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20473772

RESUMO

We tested the hypothesis that stabilizing α-helix of Epstein-Barr virus gH-derived peptide 11438 used for binding human cells will increase its biological activity. Non-stable α-helix of peptide 11438 was unfolded in an entropy-driven process, despite the opposing effect of the enthalpy factor. Adding and/or changing amino acids in peptide 11438 allowed the designing of peptides 33207, 33208 and 33210; peptides 33208 and 33210 displayed higher helical content due to a decreased unfolding entropy change as was determined by AGADIR, molecular dynamics and circular dichroism analysis. Peptides 33207, 33208 and 33210 inhibited EBV invasion of peripheral blood mononuclear cells and displayed epitopes more similar to native protein than peptide 11438; these peptides could be useful for detecting antibodies induced by native gH protein since they displayed high reactivity with anti-EBV antibodies. Anti-peptide 33207 antibodies showed higher reactivity with EBV than anti-peptide 11438 antibodies being useful for inducing antibodies against EBV. Anti-peptide 33210 antibodies inhibit EBV invasion of epithelial cells better than anti-peptide 11438 antibodies. Peptide 33210 bound to normal T lymphocytes and Raji cells stronger than peptide 11438 and also induced apoptosis of monocytes and Raji cells but not of normal T cells in a similar way to EBV-gH. Peptide 33210 inhibited the monocytes' development toward dendritic cells better than EBV and peptide 11438. In conclusion, stabilizing the α-helix in peptides 33208 and 33210 designed from peptide 11438 increased the antigenicity and the ability of the antibodies induced by peptides of inhibiting EBV invasion of host cells.


Assuntos
Antígenos Virais/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Herpesvirus Humano 4/química , Leucócitos Mononucleares/efeitos dos fármacos , Peptídeos/imunologia , Peptídeos/farmacologia , Proteínas Virais/química , Animais , Antígenos Virais/química , Antígenos Virais/genética , Linhagem Celular , Herpesvirus Humano 4/imunologia , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Testes de Sensibilidade Microbiana , Modelos Químicos , Simulação de Dinâmica Molecular , Peptídeos/síntese química , Peptídeos/química , Estrutura Secundária de Proteína , Coelhos , Termodinâmica , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...