Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Thromb Haemost ; 122(7): 1139-1146, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35052006

RESUMO

The GNE gene encodes an enzyme that initiates and regulates the biosynthesis of N-acetylneuraminic acid, a precursor of sialic acids. GNE mutations are classically associated with Nonaka myopathy and sialuria, following an autosomal recessive and autosomal dominant inheritance pattern. Reports show that single GNE variants cause severe thrombocytopenia without muscle weakness. Using panel sequencing, we identified two novel compound heterozygous variants in GNE in a young girl with life-threatening bleedings, severe congenital thrombocytopenia, and a platelet secretion defect. Both variants are located in the nucleotide-binding site of the N-acetylmannosamin kinase domain of GNE. Lectin array showed decreased α-2,3-sialylation on platelets, consistent with loss of sialic acid synthesis and indicative of rapid platelet clearance. Hematopoietic stem cell transplantation (HSCT) normalized platelet counts. This is the first report of an HSCT in a patient with an inherited GNE defect leading to normal platelet counts.


Assuntos
Miopatias Distais , Trombocitopenia , Plaquetas , Miopatias Distais/genética , Feminino , Humanos , Complexos Multienzimáticos/química , Complexos Multienzimáticos/genética , Mutação , Ácido N-Acetilneuramínico , Trombocitopenia/genética
2.
J Biol Chem ; 297(5): 101318, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34678312

RESUMO

Studying the tight activity regulation of platelet-specific integrin αIIbß3 is foundational and paramount to our understanding of integrin structure and activation. αIIbß3 is essential for the aggregation and adhesion function of platelets in hemostasis and thrombosis. Structural and mutagenesis studies have previously revealed the critical role of αIIbß3 transmembrane (TM) association in maintaining the inactive state. Gain-of-function TM mutations were identified and shown to destabilize the TM association leading to integrin activation. Studies using isolated TM peptides have suggested an altered membrane embedding of the ß3 TM α-helix coupled with αIIbß3 activation. However, controversies remain as to whether and how the TM α-helices change their topologies in the context of full-length integrin in native cell membrane. In this study, we utilized proline scanning mutagenesis and cysteine scanning accessibility assays to analyze the structure and function correlation of the αIIbß3 TM domain. Our identification of loss-of-function proline mutations in the TM domain suggests the requirement of a continuous TM α-helical structure in transmitting activation signals bidirectionally across the cell membrane, characterized by the inside-out activation for ligand binding and the outside-in signaling for cell spreading. Similar results were found for αLß2 and α5ß1 TM domains, suggesting a generalizable mechanism. We also detected a topology change of ß3 TM α-helix within the cell membrane, but only under conditions of cell adhesion and the absence of αIIb association. Our data demonstrate the importance of studying the structure and function of the integrin TM domain in the native cell membrane.


Assuntos
Membrana Celular , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Transdução de Sinais , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Células HEK293 , Humanos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Conformação Proteica em alfa-Hélice , Domínios Proteicos
3.
Proc Natl Acad Sci U S A ; 117(7): 3748-3758, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32015106

RESUMO

Increased expression of extracellular matrix (ECM) proteins in circulating tumor cells (CTCs) suggests potential function of cancer cell-produced ECM in initiation of cancer cell colonization. Here, we showed that collagen and heat shock protein 47 (Hsp47), a chaperone facilitating collagen secretion and deposition, were highly expressed during the epithelial-mesenchymal transition (EMT) and in CTCs. Hsp47 expression induced mesenchymal phenotypes in mammary epithelial cells (MECs), enhanced platelet recruitment, and promoted lung retention and colonization of cancer cells. Platelet depletion in vivo abolished Hsp47-induced cancer cell retention in the lung, suggesting that Hsp47 promotes cancer cell colonization by enhancing cancer cell-platelet interaction. Using rescue experiments and functional blocking antibodies, we identified type I collagen as the key mediator of Hsp47-induced cancer cell-platelet interaction. We also found that Hsp47-dependent collagen deposition and platelet recruitment facilitated cancer cell clustering and extravasation in vitro. By analyzing DNA/RNA sequencing data generated from human breast cancer tissues, we showed that gene amplification and increased expression of Hsp47 were associated with cancer metastasis. These results suggest that targeting the Hsp47/collagen axis is a promising strategy to block cancer cell-platelet interaction and cancer colonization in secondary organs.


Assuntos
Plaquetas/metabolismo , Neoplasias da Mama/metabolismo , Colágeno/metabolismo , Proteínas de Choque Térmico HSP47/metabolismo , Células Neoplásicas Circulantes/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Transição Epitelial-Mesenquimal , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Feminino , Amplificação de Genes , Proteínas de Choque Térmico HSP47/genética , Humanos , Camundongos SCID , Metástase Neoplásica
4.
Blood Adv ; 3(14): 2082-2092, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31296496

RESUMO

Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality in allogeneic hematopoietic stem cell transplantation (alloSCT). By static microscopy, cutaneous GVHD lesions contain a mix of T cells and myeloid cells. We used 2-photon intravital microscopy to investigate the dynamics of CD4+ and CD8+ T cells and donor dendritic cells (DCs) in cutaneous GVHD lesions in an MHC-matched, multiple minor histocompatibility antigen-mismatched (miHA) model. The majority of CD4 and CD8 cells were stationary, and few cells entered and stopped or were stopped and left the imaged volumes. CD8 cells made TCR:MHCI-dependent interactions with CD11c+ cells, as measured by the durations that CD8 cells contacted MHCI+ vs MHCI- DCs. The acute deletion of Langerin+CD103+ DCs, which were relatively rare, did not affect CD8 cell motility and DC contact times, indicating that Langerin-CD103- DCs provide stop signals to CD8 cells. CD4 cells, in contrast, had similar contact durations with MHCII+ and MHCII- DCs. However, CD4 motility rapidly increased after the infusion of an MHCII-blocking antibody, indicating that TCR signaling actively suppressed CD4 movements. Many CD4 cells still were stationary after anti-MHCII antibody infusion, suggesting CD4 cell heterogeneity within the lesion. These data support a model of local GVHD maintenance within target tissues.


Assuntos
Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/metabolismo , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Dermatopatias/etiologia , Dermatopatias/metabolismo , Linfócitos T/imunologia , Animais , Biomarcadores , Antígeno CD11c/metabolismo , Comunicação Celular , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Imunofluorescência , Expressão Gênica , Genes Reporter , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunofenotipagem , Depleção Linfocítica , Camundongos , Camundongos Transgênicos , Ligação Proteica , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T/metabolismo , Transplante Homólogo
5.
Cancer Res ; 75(8): 1580-91, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25744716

RESUMO

The extracellular matrix (ECM) is a determining factor in the tumor microenvironment that restrains or promotes malignant growth. In this report, we show how the molecular chaperone protein Hsp47 functions as a nodal hub in regulating an ECM gene transcription network. A transcription network analysis showed that Hsp47 expression was activated during breast cancer development and progression. Hsp47 silencing reprogrammed human breast cancer cells to form growth-arrested and/or noninvasive structures in 3D cultures, and to limit tumor growth in xenograft assays by reducing deposition of collagen and fibronectin. Coexpression network analysis also showed that levels of microRNA(miR)-29b and -29c were inversely correlated with expression of Hsp47 and ECM network genes in human breast cancer tissues. We found that miR-29 repressed expression of Hsp47 along with multiple ECM network genes. Ectopic expression of miR-29b suppressed malignant phenotypes of breast cancer cells in 3D culture. Clinically, increased expression of Hsp47 and reduced levels of miR-29b and -29c were associated with poor survival outcomes in breast cancer patients. Our results show that Hsp47 is regulated by miR-29 during breast cancer development and progression, and that increased Hsp47 expression promotes cancer progression in part by enhancing deposition of ECM proteins.


Assuntos
Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Matriz Extracelular/genética , Redes Reguladoras de Genes/genética , Proteínas de Choque Térmico HSP47/fisiologia , Invasividade Neoplásica/genética , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , MicroRNAs/fisiologia , Transplante de Neoplasias , Células Tumorais Cultivadas
6.
Histol Histopathol ; 29(9): 1083-92, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24682974

RESUMO

Extracellular matrix (ECM), a major component of the cellular microenvironment, plays critical roles in normal tissue morphogenesis and disease progression. Binding of ECM to membrane receptor proteins, such as integrin, discoidin domain receptors, and dystroglycan, elicits biochemical and biomechanical signals that control cellular architecture and gene expression. These ECM signals cooperate with growth factors and hormones to regulate cell migration, differentiation, and transformation. ECM signaling is tightly regulated during normal mammary gland development. Deposition and alignment of fibrillar collagens direct migration and invasion of mammary epithelial cells during branching morphogenesis. Basement membrane proteins are required for polarized acinar morphogenesis and milk protein expression. Deregulation of ECM proteins in the long run is sufficient to promote breast cancer development and progression. Recent studies demonstrate that the integrated biophysical and biochemical signals from ECM and soluble factors are crucial for normal mammary gland development as well as breast cancer progression.


Assuntos
Neoplasias da Mama/metabolismo , Matriz Extracelular/metabolismo , Glândulas Mamárias Humanas/embriologia , Glândulas Mamárias Humanas/metabolismo , Neoplasias da Mama/patologia , Progressão da Doença , Matriz Extracelular/patologia , Feminino , Humanos , Transdução de Sinais/fisiologia
7.
BMC Cancer ; 14: 1, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24383403

RESUMO

BACKGROUND: Increased collagen deposition provides physical and biochemical signals to support tumor growth and invasion during breast cancer development. Therefore, inhibition of collagen synthesis and deposition has been considered a strategy to suppress breast cancer progression. Collagen prolyl-4-hydroxylase α subunit 2 (P4HA2), an enzyme hydroxylating proline residues in -X-Pro-Gly- sequences, is a potential therapeutic target for the disorders associated with increased collagen deposition. However, expression and function of P4HA2 in breast cancer progression are not well investigated. METHODS: Gene co-expression analysis was performed in the published microarray datasets to identify potential regulators of collagen I, III, and IV in human breast cancer tissue. Expression of P4HA2 was silenced by shRNAs, and its activity was inhibited by 1, 4-DPCA, a prolyl-4-hydroxylase inhibitor. Three-dimensional culture assay was used to analyze roles of P4HA2 in regulating malignant phenotypes of breast cancer cells. Reduced deposition of collagen I and IV was detected by Western blotting and immunofluorescence. Control and P4HA2-silenced breast cancer cells were injected into fat pad and tail vein of SCID mice to examine effect of P4HA2 on tumor growth and lung metastasis. RESULTS: Using gene co-expression analysis, we showed that P4HA2 was associated with expression of Col1A1, Col3A1, and Col4A1 during breast cancer development and progression. P4HA2 mRNA levels were significantly upregulated in breast cancer compared to normal mammary tissue. Increased mRNA levels of P4HA2 correlated with poor clinical outcome in breast cancer patients, which is independent of estrogen receptor status. Silencing P4HA2 expression or treatment with the P4HA inhibitor significantly inhibited cell proliferation and suppressed aggressive phenotypes of breast cancer cells in 3D culture, accompanied by reduced deposition of collagen I and IV. We also found that knockdown of P4HA2 inhibited mammary tumor growth and metastasis to lungs in xenograft models. CONCLUSION: These results suggest the critical role of P4HA2 in breast cancer progression and identify P4HA2 as a potential therapeutic target and biomarker for breast cancer progression.


Assuntos
Neoplasias da Mama/enzimologia , Colágeno/metabolismo , Neoplasias Pulmonares/enzimologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colágeno/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Invasividade Neoplásica , Fenótipo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , Prognóstico , Prolil Hidroxilases/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Immunology ; 141(2): 211-21, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24117005

RESUMO

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptor (TRAIL-R) play important roles in immune regulation and cancer cell death. Although TRAIL has been shown to induce chemokine release in various tumour cells, the function of TRAIL-R in the development of colitis and colitis-associated carcinogenesis has not been explored. In this study, we found that TRAIL-R-deficient mice exhibited a higher incidence of colitis and colitis-associated cancer than that of wild-type (WT) mice, and TRAIL-R expression was down-regulated in WT mice that were fed dextran sulphate sodium. Chemokines, including CCL2 and CXCL1, were highly expressed in the serum and inflammatory colon tissues of TRAIL-R(-/-) mice compared with WT mice, and TRAIL-R(-/-) mice showed a marked infiltration of immune cells during colitis. Hyperactivation of Janus kinase and nuclear factor-κB in colon epithelial cells was also observed, which correlated with the severity of colonic inflammation in TRAIL-R(-/-) mice. These data suggest that TRAIL-R plays a protective role in chemical-induced colon injury and negatively regulates mucosal immune responses.


Assuntos
Colite/prevenção & controle , Neoplasias do Colo/etiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Animais , Movimento Celular , Quimiocina CCL2/análise , Quimiocina CXCL1/análise , Colite/induzido quimicamente , Colite/complicações , Sulfato de Dextrana , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/deficiência
9.
Cancer Immunol Immunother ; 62(4): 653-63, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23143747

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a promising and novel anticancer cytokine, specifically kills numerous tumor cells by apoptosis. However, some malignancies are resistant to TRAIL treatment in clinical trials, thus limiting its therapeutic potential. In the present study, the TRAIL-resistant murine hepatocellular carcinoma cell line Hepa1-6 was used to elucidate the physiological significance of TRAIL resistance, especially with respect to the immune regulatory function of TRAIL. Hepa1-6 cells were resistant to TRAIL-induced apoptosis in vitro; however, intratumoral injection of recombinant soluble TRAIL inhibited tumor growth and prolonged survival time in tumor-bearing mice. Local TRAIL treatment decreased the number of intratumoral CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) but did not affect CD4(+)CD25(+)Foxp3(+) Tregs in the draining lymph nodes and spleen. Further investigation showed that TRAIL induced apoptosis of tumor-activated CD4(+)CD25(+)Foxp3(+) Tregs, but not of CD4(+)CD25(-) T cells. Moreover, mouse TRAIL receptor DR5 expression was detected on the surface of the tumor-infiltrating CD4(+)CD25(+)Foxp3(+) Tregs, but not on naïve CD4(+)CD25(+)Foxp3(+) Tregs. Interestingly, intratumoral injection of TRAIL not only decreased the number of CD4(+)CD25(+)Foxp3(+) Tregs but also increased the number of tumor-specific CD8(+) CTL and augmented their cytotoxicity to the tumor cells. These data provide the novel evidence for an immune regulatory function of TRAIL and may shed light on the clinical application of TRAIL.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Hepáticas Experimentais/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Subunidade alfa de Receptor de Interleucina-2/imunologia , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia
10.
Blood ; 119(26): 6317-25, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22490676

RESUMO

Arginine-glycine-aspartic acid (RGD)-mimetic platelet inhibitors act by occupying the RGD recognition site of α(IIb)/ß(3) integrin (GPIIb/IIIa), thereby preventing the activated integrin from reacting with fibrinogen. Thrombocytopenia is a well-known side effect of treatment with this class of drugs and is caused by Abs, often naturally occurring, that recognize α(IIb)/ß(3) in a complex with the drug being administered. RGD peptide and RGD-mimetic drugs are known to induce epitopes (ligand-induced binding sites [LIBS]) in α(IIb)/ß(3) that are recognized by certain mAbs. It has been speculated, but not shown experimentally, that Abs from patients who develop thrombocytopenia when treated with an RGD-mimetic inhibitor similarly recognize LIBS determinants. We addressed this question by comparing the reactions of patient Abs and LIBS-specific mAbs against α(IIb)/ß(3) in a complex with RGD and RGD-mimetic drugs, and by examining the ability of selected non-LIBS mAbs to block binding of patient Abs to the liganded integrin. Findings made provide evidence that the patient Abs recognize subtle, drug-induced structural changes in the integrin head region that are clustered about the RGD recognition site. The target epitopes differ from classic LIBS determinants, however, both in their location and by virtue of being largely drug-specific.


Assuntos
Anticorpos/efeitos adversos , Anticorpos/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/imunologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombocitopenia/etiologia , Animais , Anticorpos/imunologia , Células CHO , Cricetinae , Cricetulus , Eptifibatida , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Oligopeptídeos/química , Oligopeptídeos/imunologia , Peptídeos/uso terapêutico , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/uso terapêutico , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Ligação Proteica/imunologia , Conformação Proteica , Ratos , Especificidade por Substrato , Trombocitopenia/tratamento farmacológico , Trombocitopenia/imunologia , Tirofibana , Tirosina/análogos & derivados , Tirosina/uso terapêutico
11.
Blood ; 120(2): 449-58, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22490677

RESUMO

In the present study, we re-annotated von Willebrand factor (VWF), assigned its entire sequence to specific modules, and related these modules to structure using electron microscopy (EM). The D domains are assemblies of smaller modules visible as lobes in EM. Modules in the D-domain assemblies include von Willebrand D, 8-cysteine, trypsin inhibitor-like, E or fibronectin type 1-like domains, and a unique D4N module in D4. The D1-D2 prodomain shows 2 large connected assemblies, each containing smaller lobes. The previous B and C regions of VWF are re-annotated as 6 tandem von Willebrand C (VWC) and VWC-like domains. These 6 VWC domains correspond to 6 elongated domains that associate in pairs at acidic pH in the stem region of VWF dimeric bouquets. This correspondence is demonstrated by binding of integrin α(IIb)ß(3) to the fourth module seen in EM, VWC4, which bears the VWF Arg-Gly-Asp motif. The C-terminal cystine knot domain dimerizes end-to-end in a manner predicted by homology to TGF-ß and orients approximately perpendicular to the VWC domains in dimeric bouquets. Homologies of domains in VWF to domains in other proteins allow many disulfide bonds to be tentatively assigned, which may have functional implications.


Assuntos
Fator de von Willebrand/química , Fator de von Willebrand/genética , Sequência de Aminoácidos , Sítios de Ligação , Dimerização , Humanos , Microscopia Eletrônica , Modelos Moleculares , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos , Fator de von Willebrand/ultraestrutura
12.
Arthritis Rheum ; 64(5): 1345-54, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22131069

RESUMO

OBJECTIVE: Synovial cells in rheumatoid synovium display abnormal proliferation, which leads to joint destruction. TRAIL has been described as a proapoptotic factor in fibroblast-like synoviocytes (FLS). This study was undertaken to investigate the functions of rAAV2/5-TRAIL in human FLS and in arthritic mice. METHODS: Primary human FLS were infected with rAAV2/5-TRAIL in the presence or absence of epirubicin. Transgene expression was monitored by both enzyme-linked immunosorbent assay and flow cytometry. The disease-modulating activity of epirubicin plus rAAV2/5-TRAIL was investigated in mice with collagen-induced arthritis (CIA). RESULTS: Subtoxic doses of epirubicin potentiated rAAV2/5-mediated TRAIL expression in FLS and simultaneously enhanced the sensitivity of FLS to TRAIL. Epirubicin treatment up-regulated death receptor 4 (DR-4) and DR-5 expression and down-regulated FLIP expression, thereby enhancing the activation of procaspase 3, procaspase 8, and procaspase 9. An in vivo study showed that the combination of rAAV2/5-TRAIL gene therapy and epirubicin chemotherapy provided augmented antiarthritic effects in a mouse model of CIA. The intraarticular injection of rAAV2/5-TRAIL combined with epirubicin treatment significantly reduced the severity and incidence of CIA and joint swelling in the animals. Histologic evaluations revealed that inflammatory cell infiltration, cartilage destruction, and bone erosion were significantly reduced in the joints of the mice receiving the synthetic treatment. Results of a viral genome copy number assay indicated that epirubicin dramatically augmented the expression of rAAV2/5-TRAIL without altering its tissue distribution. CONCLUSION: These results suggest that epirubicin enhances the antiarthritic effect of rAAV2/5-TRAIL and that combination treatment might be an important therapeutic alternative, with practical significance for rheumatoid arthritis.


Assuntos
Adenoviridae/genética , Antibióticos Antineoplásicos/uso terapêutico , Artrite Experimental/terapia , Epirubicina/farmacologia , Terapia Genética/métodos , Membrana Sinovial/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/genética , Adenoviridae/classificação , Adenoviridae/imunologia , Adulto , Animais , Anticorpos Antivirais/biossíntese , Artrite Experimental/genética , Artrite Experimental/patologia , Terapia Combinada , Modelos Animais de Doenças , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Marcação de Genes/métodos , Vetores Genéticos/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Pessoa de Meia-Idade , Proteínas Recombinantes , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
13.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 33(4): 367-70, 2011 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-21906442

RESUMO

OBJECTIVE: To investigate the mechanism of anti-death receptor 5-10 (AD5-10) combined with epirubicin in treating rheumatoid arthritis (RA). METHODS: We detected the cell viability of the fibroblast-like synoviocytes (FLS) from RA patients with MTT. The expression level of apoptosis signaling pathways protein, p53, and p21 were evaluated with Western blot. RESULTS: We found that epirubicin, at different doses, could enhance the effect of AD5-10 on FLS, promoting the apoptosis of FLS. The expression levels of caspase-3, -8, -9, c-FLIP, Bcl-2, p53, and p21 in the FLS changed after epirubicin treatment. CONCLUSION: Epirubicin may coordinate with AD5-10 in inducing FLS apoptosis through affecting the levels of p53, p21, c-FLIP, and Bcl-2.


Assuntos
Anticorpos Monoclonais/farmacologia , Epirubicina/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Membrana Sinovial/citologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Humanos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo
14.
J Biol Chem ; 286(3): 1860-7, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21081497

RESUMO

Integrin transmembrane (TM) and/or cytoplasmic domains play a critical role in integrin bidirectional signaling. Although it has been shown that TM and/or cytoplasmic α and ß domains associate in the resting state and separation of these domains is required for both inside-out and outside-in signaling, the role of TM homomeric association remains elusive. Formation of TM homo-oligomers was observed in micelles and bacterial membranes previously, and it has been proposed that homomeric association is important for integrin activation and clustering. This study addresses whether integrin TM domains form homo-oligomers in mammalian cell membranes using cysteine scanning mutagenesis. Our results show that TM homomeric interaction does not occur before or after soluble ligand binding or during inside-out activation. In addition, even though the cysteine mutants and the heterodimeric disulfide-bounded mutant could form clusters after adhering to immobilized ligand, the integrin TM domains do not form homo-oligomers, suggesting that integrin TM homomeric association is not critical for integrin clustering or outside-in signaling. Therefore, integrin TM homo-oligomerization is not required for integrin activation, ligand binding, or signaling.


Assuntos
Integrinas/metabolismo , Multimerização Proteica/fisiologia , Transdução de Sinais/fisiologia , Substituição de Aminoácidos , Adesão Celular , Linhagem Celular , Humanos , Integrinas/genética , Ligantes , Mutagênese , Ligação Proteica , Estrutura Terciária de Proteína
15.
Mol Cell Biol ; 30(22): 5432-43, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20837704

RESUMO

The mechanisms by which signals are transmitted across the plasma membrane to regulate signaling are largely unknown for receptors with single-pass transmembrane domains such as the epidermal growth factor receptor (EGFR). A crystal structure of the extracellular domain of EGFR dimerized by epidermal growth factor (EGF) reveals the extended, rod-like domain IV and a small, hydrophobic domain IV interface compatible with flexibility. The crystal structure and disulfide cross-linking suggest that the 7-residue linker between the extracellular and transmembrane domains is flexible. Disulfide cross-linking of the transmembrane domain shows that EGF stimulates only moderate association in the first two α-helical turns, in contrast to association throughout the membrane over five α-helical turns in glycophorin A and integrin. Furthermore, systematic mutagenesis to leucine and phenylalanine suggests that no specific transmembrane interfaces are required for EGFR kinase activation. These results suggest that linkage between ligand-induced dimerization and tyrosine kinase activation is much looser than was previously envisioned.


Assuntos
Receptores ErbB/química , Receptores ErbB/metabolismo , Ligantes , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Reagentes de Ligações Cruzadas/química , Cisteína/química , Cisteína/genética , Dimerização , Dissulfetos/química , Ativação Enzimática , Receptores ErbB/genética , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência , Difração de Raios X
16.
Biochem Biophys Res Commun ; 348(3): 916-22, 2006 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-16904649

RESUMO

The fusion proteins of enveloped viruses mediating the fusion between the viral and cellular membranes comprise two discontinuous heptad repeat (HR) domains located at the ectodomain of the enveloped glycoproteins. The crystal structure of the fusion protein core of Mumps virus (MuV) was determined at 2.2 A resolution. The complex is a six-helix bundle in which three HR1 peptides form a central highly hydrophobic coiled-coil and three HR2 peptides pack against the hydrophobic grooves on the surface of central coiled-coil in an oblique antiparallel manner. Fusion core of MuV, like those of simian virus 5 and human respiratory syncytium virus, forms typical 3-4-4-4-3 spacing. The similar characterization in HR1 regions, as well as the existence of O-X-O motif in extended regions of HR2 helix, suggests a basic rule for the formation of the fusion core of viral fusion proteins.


Assuntos
Fusão de Membrana/fisiologia , Vírus da Caxumba/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Sequência Conservada , Cristalografia por Raios X , Dados de Sequência Molecular , Vírus da Caxumba/patogenicidade , Vírus da Parainfluenza 5/química , Vírus da Parainfluenza 5/patogenicidade , Fragmentos de Peptídeos/química , Conformação Proteica , Estrutura Secundária de Proteína , Sequências Repetitivas de Aminoácidos , Proteínas Virais de Fusão/fisiologia
17.
Antivir Ther ; 10(7): 833-40, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16312179

RESUMO

Human respiratory syncytial virus (hRSV) is a pathogen of worldwide health concern. The crucial membrane fusion event during viral entry into host cells involves a 'trimer-of-hairpins' structure that brings the amino (N)- and carboxy (C)-terminal regions of the viral fusion glycoprotein (F protein) into close proximity. Two heptad repeat regions that are highly conserved in the F protein - HR1 (N-terminal) and HR2 (C-terminal) - have an important role in this process. It has been shown that both HR1-and HR2-based peptides can inhibit viral entry. However, these proteins, and the HR1 peptides in particular, are liable to aggregation. We designed three peptides containing multiple copies of alternating HR1 and HR2 sequences denoted 5-Helix, HR121 and HR212, respectively. The 5-Helix, HR121 and HR212 proteins were functionally analogous to single HR1, HR1 and HR2 sequences, respectively. All three proteins were expressed in soluble form and biophysical analysis showed that they exhibited alpha-helical secondary structures. The three proteins were potent fusion inhibitors in vitro, at the micromolar scale, with the HR1 analogues being approximately two times more effective than the HR2 analogue. Our results suggest that these rationally designed protein inhibitors could serve as a new class of anti-hRSV agents.


Assuntos
Antivirais/química , Antivirais/farmacologia , Desenho de Fármacos , Fusão de Membrana/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/efeitos dos fármacos , Vírus Sincicial Respiratório Humano/fisiologia , Proteínas Virais de Fusão/antagonistas & inibidores , Linhagem Celular Tumoral , Membrana Celular/virologia , Relação Dose-Resposta a Droga , Humanos , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Infecções por Vírus Respiratório Sincicial/virologia , Proteínas Virais de Fusão/metabolismo
18.
J Mol Biol ; 354(3): 601-13, 2005 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-16253271

RESUMO

Paramyxovirus infections can be detected worldwide with some emerging zoonotic viruses and currently there are no specific therapeutic treatments or vaccines available for many of these diseases. Recent studies have demonstrated that peptides derived from the two heptad repeat regions (HR1 and HR2) of paramyxovirus fusion proteins could be used as inhibitors of virus fusion. The mechanism underlying this activity is in accordance with that of class I virus fusion proteins, of which human immunodeficiency virus (HIV) and influenza virus fusion proteins are members. For class I virus fusion proteins, the HR1 fragment binds to HR2 to form a six-helix bundle with three HR1 fragments forming the central coiled bundle surrounded by three coiled HR2 fragments in the post fusion conformational state (fusion core). It is hypothesized that the introduced exogenous HR1 or HR2 can compete against their endogenous counterparts, which results in fusion inhibition. Using Newcastle disease virus (NDV) as a model, we designed several protein inhibitors, denoted HR212 as well asHR121 and 5-Helix, which could bind the HR1 or HR2 region of fusion protein, respectively. All the proteins were expressed and purified using a GST-fusion expression system in Escherichia coli. The HR212 or GST-HR212 protein, which binds the HR1 peptide in vitro, displayed inhibitory activity against NDV-mediated cell fusion, while the HR121 and 5-Helix proteins, which bind the HR2 peptide in vitro, inhibited virus fusion from the avirulent NDV strain when added before the cleavage of the fusion protein. These results showed that the designed HR212, HR121 or 5-Helix protein could serve as specific antiviral agents. These data provide additional insight into the difference between the virulent and avirulent strains of NDV.


Assuntos
Desenho de Fármacos , Fusão de Membrana/efeitos dos fármacos , Vírus da Doença de Newcastle/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Cromatografia em Gel , Dicroísmo Circular , Células HeLa , Humanos , Fusão de Membrana/fisiologia , Dados de Sequência Molecular , Vírus da Doença de Newcastle/química , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Fragmentos de Peptídeos/genética , Ligação Proteica , Desnaturação Proteica , Estrutura Secundária de Proteína , Solubilidade , Temperatura , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética
19.
Biochem Biophys Res Commun ; 329(2): 603-9, 2005 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-15737628

RESUMO

Entry of lentiviruses, such as human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV), requires folding of two heptad repeat regions (HR1 and HR2) of gp41 into a trimer-of-hairpins, which subsequently brings virus and cell membrane into fusion. This motif is a generalized feature of viral fusion proteins and has been exploited in generating antiviral fusion agents. In the present paper, we report structural characters of Env protein from another lentivirus, bovine immunodeficiency virus (BIV), which contributes to a good animal model of HIV. BIV HR1 and HR2 regions are predicted by two different programs and expressed separately or conjointly in Escherichia coli. Biochemical and biophysical analyses show that the predicted HRs of BIV Env can form a stable trimer-of-hairpins or six-helix bundle just like that formed by feline immunodeficiency virus Env. Cell fusion assay demonstrates that the HR2 peptide of BIV can efficiently inhibit the virus-mediated cell fusion.


Assuntos
Vírus da Imunodeficiência Bovina/fisiologia , Pulmão/virologia , Fusão de Membrana/fisiologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Fusão Celular , Células Cultivadas , Pulmão/citologia , Conformação Molecular , Dados de Sequência Molecular , Peso Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
20.
Biochem Biophys Res Commun ; 330(1): 39-45, 2005 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15781229

RESUMO

Entry of SARS-CoV into a target cell is initiated by binding of the S1 domain of spike protein to a receptor, followed by conformational changes of the spike protein S2 domain, resulting in the formation of a six-helix bundle by the heptad-repeat (HR1 and HR2) regions. Our previous studies have demonstrated that peptides derived from HR2 region could inhibit SARS-CoV entry. However, synthesis of these peptides is at high cost. In this study, we designed two recombinant proteins, one containing two HR1 and one HR2 peptides (denoted HR121), and the other consisting of two HR2 and one HR1 peptides (designated HR212). These two proteins could be easily purified with the low cost of production, exhibiting high stability and potent inhibitory activity on entry of the HIV/SARS pseudoviruses with IC(50) values of 4.13 and 0.95muM, respectively. These features suggest that HR121 and HR212 can serve as potent inhibitors of SARS-CoV entry.


Assuntos
Fusão de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/antagonistas & inibidores , Proteínas Recombinantes/farmacologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Proteínas do Envelope Viral/antagonistas & inibidores , Linhagem Celular , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Humanos , Glicoproteína da Espícula de Coronavírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA