Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Ann Clin Lab Sci ; 52(2): 170-178, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35414495

RESUMO

OBJECTIVE: Found in Inflammatory Zone 1 (FIZZ1) protein plays an important enhancive role in inflammation and angiogenesis. This study aims to explore the effects of FIZZ1 on murine atherosclerosis. METHODS: The murine aortic endothelial cells were treated with an adenoviral vector encoding FIZZ1 short hairpin RNA (Ad-shFIZZ1). Murine atherosclerosis model was established with ApoE-/- mice. The effects of FIZZ1 were studied in vitro and in vivo. RESULTS: Ad-shFIZZ1 treatment suppressed the expression of FIZZ1 and the formation of capillary tube formation in vitro. Administration of Ad-shFIZZ1 suppressed FIZZ1-mediated signaling, the growth of endothelial cells and the production of inflammatory molecules in murine aorta in vivo, and inhibited the development of atherosclerosis in vivo. CONCLUSION: FIZZ1 played a potent promotive role in atherosclerosis, and could serve as a novel therapeutic target for the treatment of the disease.


Assuntos
Aterosclerose , Células Endoteliais , Animais , Aterosclerose/genética , Células Endoteliais/metabolismo , Humanos , Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/genética
2.
Aging (Albany NY) ; 11(22): 10016-10030, 2019 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-31757932

RESUMO

This study aimed to explore the interactions among long non-coding RNA H19, transcriptional factor CCCTC-binding factor (CTCF) and polycystic kidney disease 1 (PKD1), and to investigate its potentially regulatory effect on vulnerable plaque formation and angiogenesis of atherosclerosis. We established an atherosclerosis mouse model in ApoE knockout mice, followed by gain- and loss-of-function approaches. H19 was upregulated in aortic tissues of atherosclerosis mice, but silencing of H19 significantly inhibited atherosclerotic vulnerable plaque formation and intraplaque angiogenesis, accompanied by a downregulated expression of MMP-2, VEGF, and p53 and an upregulated expression of TIMP-1. Moreover, opposite results were found in the aortic tissues of atherosclerosis mice treated with H19 or CTCF overexpression. H19 was capable of recruiting CTCF to suppress PKD1, thus promoting atherosclerotic vulnerable plaque formation and intraplaque angiogenesis in atherosclerosis mice. The present study provides evidence that H19 recruits CTCF to downregulate the expression of PKD1, thereby promoting vulnerable plaque formation and intraplaque angiogenesis in mice with atherosclerosis.


Assuntos
Aorta/metabolismo , Aterosclerose/metabolismo , Fator de Ligação a CCCTC/metabolismo , Regulação para Baixo , Proteína Quinase C/metabolismo , RNA Longo não Codificante/metabolismo , Regulação para Cima , Animais , Aterosclerose/genética , Fator de Ligação a CCCTC/genética , Modelos Animais de Doenças , Inativação Gênica , Camundongos , Camundongos Knockout para ApoE , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Proteína Quinase C/genética , RNA Longo não Codificante/genética
3.
Biomed Rep ; 1(1): 1-5, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31258903

RESUMO

Left ventricular hypertrophy is a leading cause of heart failure and sudden death. Cysteine-rich transmembrane bone morphogenetic protein regulator 1 (Crim1) is expressed at a high level in the heart and has a regulatory role in heart development. The present study aimed to test the hypothesis that Crim1 can have an inhibitory function on ventricular hypertrophy. Rat primary ventricular myocytes were stretched to induce myocyte hypertrophy, and treated with telmisartan or infected with Crim1-expressing recombinant adenovirus (Ad-Crim1). Rat ventricular hypertrophy was induced by abdominal aortic coarctation (AAC), and treated either with telmisartan or myocardial injection of Ad-Crim1 or empty adenovirus vector. The results showed that the expression of Crim1 decreased in the hypertrophic ventricle. The inhibition of angiotensin receptor type 1 (AT1R) by telmisartan in vitro and in vivo significantly increased the expression of Crim1 in the left ventricle. The overexpression of Crim1 by infection with Ad-Crim1 significantly inhibited stretch-induced ventricular myocyte hypertrophy in vitro. The overexpression of Crim1 by gavage with AT1R inhibitor telmisartan or myocardial injection of Ad-Crim1 markedly suppressed AAC-induced left ventricular hypertrophy in vivo. These results suggest that Crim1 has a suppressive function on ventricular hypertrophy and provides a novel therapeutic target for the treatment of cardiac hypertrophy.

4.
Lipids Health Dis ; 17(1): 261, 2018 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-30458796

RESUMO

BACKGROUND: Colon cancer is a malignancy of the large intestine with high mortality and economic burden. Recent studies reveal a new relationship between blood lipids and the risk of cancer. The presents study aims to investigate the combination of serum lipids with cancer antigens as a novel diagnostic marker for colon cancer. METHODS: Two hundred of colon cancer patients or healthy subjects were recruited. Serum lipids and cancer antigens such as total cholesterol (TC), high-density lipoprotein (HDL), carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA19-9) were measured. RESULTS: There were significantly lower level of serum TC or HDL, and significantly higher level of serum CEA or CA19-9 in patients than in healthy subjects. Serum TC or HDL in patients with advanced colon cancer was significantly lower than the ones with early stage disease. The level of serum TC or HDL in patients after surgical removal of colon cancer was significantly higher compared to the ones before surgery, but serum CEA or CA19-9 after surgery was significantly reduced in comparison with the ones before surgery. Combined TC, HDL, CEA and CA19-9 as a diagnostic marker for colon cancer had the highest positive predictive rate in comparison with individual, two or three of the parameters. CONCLUSIONS: The combination of serum TC, HDL, CEA and CA19-9 can be used as an effective marker for colon cancer, and offers a novel strategy for clinical diagnosis and monitoring the disease.


Assuntos
Biomarcadores Tumorais/sangue , Antígeno CA-19-9/sangue , Antígeno Carcinoembrionário/sangue , Neoplasias do Colo/sangue , Lipídeos/sangue , Humanos , Lipoproteínas HDL/sangue
5.
Oncogene ; 37(25): 3415-3425, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29551769

RESUMO

Monocyte chemotactic protein-induced protein-1 (MCPIP1; also called Regnase-1) encoded by the ZC3H12A gene critically regulates inflammatory responses and immune homeostasis primarily by RNase-dependent and -independent mechanisms. However, the relationship of MCPIP1 with apoptosis and cancer and the underlying mechanisms are largely unclear. The current study has demonstrated a previously uncovered connection between MCPIP1 and the negative regulation of death receptor 5 (DR5; also known as TRAIL-R2 or killer/DR5), a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is produced endogenously by various immune cells such as T cells. Our findings have revealed that MCPIP1 decreases both total cellular and cell surface DR5, primarily through modulating DUB-mediated protein autophagic/lysosomal degradation. Suppression of MCPIP1 by gene knockdown induces the formation of death-induced signaling complex (DISC) and enhances TRAIL or DR5 activation-induced apoptosis in cancer cells. Moreover, we demonstrated an inverse correlation between MCPIP1 expression and DR5 expression/cell sensitivity to DR5 activation-induced apoptosis in cancer cells. Our findings warrant future investigation of the roles of negative regulation of DR5 by MCPIP1 in cancer and in T-cell immunity.


Assuntos
Apoptose , Neoplasias da Mama/patologia , Enzimas Desubiquitinantes/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ribonucleases/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , Enzimas Desubiquitinantes/genética , Feminino , Humanos , Proteólise , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ribonucleases/genética , Transdução de Sinais , Fatores de Transcrição/genética , Células Tumorais Cultivadas
6.
Sci Rep ; 7(1): 8027, 2017 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-28808321

RESUMO

b-AP15 and its derivatives block proteasome deubiquitinase (DUB) activity and have been developed and tested in the clinic as potential cancer therapeutic agents. b-AP15 induces apoptosis in cancer cells, but the underlying mechanisms are largely undefined. The current study focuses on studying the modulatory effects of b-AP15 on death receptor 5 (DR5) levels and DR5 activation-induced apoptosis as well as on understanding the underlying mechanisms. Treatment with b-AP15 potently increased DR5 levels including cell surface DR5 in different cancer cell lines with limited or no effects on the levels of other related proteins including DR4, c-FLIP, FADD, and caspase-8. b-AP15 substantially slowed the degradation of DR5, suggesting that it stabilizes DR5. Moreover, b-AP15 effectively augmented apoptosis when combined with TRAIL or the DR5 agonistic antibody AMG655; these effects are DR5-dependent because DR5 deficiency abolished the ability of b-AP15 to enhance TRAIL- or AMG655-induced apoptosis. Therefore, it is clear that b-AP15, and possibly its derivatives, can stabilize DR5 and increase functional cell surface DR5 levels, resulting in enhancement of DR5 activation-induced apoptosis. Our findings suggest that b-AP15 and its derivatives may have potential in sensitizing cancer cells to DR5 activation-based cancer therapy.


Assuntos
Apoptose/efeitos dos fármacos , Piperidonas/farmacologia , Inibidores de Proteases/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Células HCT116 , Humanos , Estabilidade Proteica , Proteólise
7.
Mol Ther ; 25(2): 416-426, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28153092

RESUMO

Hematopoietic stem cells (HSCs) have the capacity to self-renew and differentiate into hematopoietic cells and have been utilized to replace diseased bone marrow for patients with cancers and blood disorders. Although remarkable progress has been made in developing new tools to manipulate HSCs for clinic use, there is still no effective method to expand HSCs in vivo for quick repopulation of hematopoietic cells following sublethal irradiation. We have recently described a novel synthetic cytokine that is derived from the fusion of granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin 4 (IL-4; named as GIFT4), and we have now discovered that GIFT4 fusokine promotes long-term hematopoietic regeneration in a B cell-dependent manner. We found that GIFT4 treatment triggered a robust expansion of endogenous bone marrow HSCs and multipotent progenitors in vivo. Delivery of GIFT4 protein together with B cells rescued lethally irradiated mice. Moreover, adoptive transfer of autologous or allogeneic GIFT4-treated B cells (GIFT4-B cells) enhanced long-term hematopoietic recovery in radiated mice and prevented the mice from irradiation-induced death. Our data suggest that GIFT4 as well as GIFT4-B cells could serve as means to augment HSC engraftment in the setting of bone marrow transplantation for patients with hematological malignancy.


Assuntos
Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Interleucina-4 , Linfopoese/efeitos dos fármacos , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linfócitos B/metabolismo , Biomarcadores , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Imunofenotipagem , Interleucina-4/genética , Masculino , Camundongos , Modelos Animais , Fenótipo , Proteínas Recombinantes de Fusão/genética
8.
J Biol Chem ; 291(41): 21694-21702, 2016 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-27576686

RESUMO

Death receptor 4 (DR4) is a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and triggers apoptosis upon ligation with TRAIL or aggregation. MEK/ERK signaling is a well known and the best-studied effector pathway downstream of Ras and Raf. This study focuses on determining the impact of pharmacological MEK inhibition on DR4 expression and elucidating the underlying mechanism. We found that several MEK inhibitors including MEK162, AZD6244, and PD0325901 effectively decreased DR4 protein levels including cell surface DR4 in different cancer cell lines. Accordingly, pre-treatment of TRAIL-sensitive cancer cell lines with a MEK inhibitor desensitized them to TRAIL-induced apoptosis. These results indicate that MEK inhibition negatively regulates DR4 expression and cell response to TRAIL-induced apoptosis. MEK inhibitors did not alter DR4 protein stability, rather decreased its mRNA levels, suggesting a transcriptional regulation. In contrast, enforced activation of MEK/ERK signaling by expressing ectopic B-Raf (V600E) or constitutively activated MEK1 (MEK1-CA) or MEK2 (MEK2-CA) activated ERK and increased DR4 expression; these effects were inhibited when a MEK inhibitor was present. Promoter analysis through deletion and mutation identified the AP-1 binding site as an essential response element for enhancing DR4 transactivation by MEK1-CA. Furthermore, inhibition of AP-1 by c-Jun knockdown abrogated the ability of MEK1-CA to increase DR4 promoter activity and DR4 expression. These results suggest an essential role of AP-1 in mediating MEK/ERK activation-induced DR4 expression. Our findings together highlight a previously undiscovered mechanism that positively regulates DR4 expression through activation of the MEK/ERK/AP-1 signaling pathway.


Assuntos
Regulação Neoplásica da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Neoplasias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Substituição de Aminoácidos , Linhagem Celular Tumoral , Células HEK293 , Humanos , Mutação de Sentido Incorreto , Neoplasias/genética , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Transcrição AP-1/genética
9.
Cancer Res ; 76(19): 5683-5695, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27488533

RESUMO

The clinical efficacy of immune cytokines used for cancer therapy is hampered by elements of the immunosuppressive tumor microenvironment such as TGFß. Here we demonstrate that FIST15, a recombinant chimeric protein composed of the T-cell-stimulatory cytokine IL15, the sushi domain of IL15Rα and a TGFß ligand trap, can overcome immunosuppressive TGFß to effectively stimulate the proliferation and activation of natural killer (NK) and CD8+ T cells with potent antitumor properties. FIST15-treated NK and CD8+ T cells produced more IFNγ and TNFα compared with treatment with IL15 and a commercially available TGFß receptor-Fc fusion protein (sTßRII) in the presence of TGFß. Murine B16 melanoma cells, which overproduce TGFß, were lysed by FIST15-treated NK cells in vitro at doses approximately 10-fold lower than NK cells treated with IL15 and sTßRII. Melanoma cells transduced to express FIST15 failed to establish tumors in vivo in immunocompetent murine hosts and could only form tumors in beige mice lacking NK cells. Mice injected with the same cells were also protected from subsequent challenge by unmodified B16 melanoma cells. Finally, mice with pre-established B16 melanoma tumors responded to FIST15 treatment more strongly compared with tumors treated with control cytokines. Taken together, our results offer a preclinical proof of concept for the use of FIST15 as a new class of biological therapeutics that can coordinately neutralize the effects of immunosuppressive TGFß in the tumor microenvironment while empowering tumor immunity. Cancer Res; 76(19); 5683-95. ©2016 AACR.


Assuntos
Interleucina-15/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Humanos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Microambiente Tumoral
10.
Sci Rep ; 6: 26803, 2016 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-27222248

RESUMO

B-Raf inhibitors have been used for the treatment of some B-Raf-mutated cancers. They effectively inhibit B-Raf/MEK/ERK signaling in cancers harboring mutant B-Raf, but paradoxically activates MEK/ERK in Ras-mutated cancers. Death receptor 5 (DR5), a cell surface pro-apoptotic protein, triggers apoptosis upon ligation with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or aggregation. This study focused on determining the effects of B-Raf inhibition on DR5 expression and DR5 activation-induced apoptosis in Ras-mutant cancer cells. Using chemical and genetic approaches, we have demonstrated that the B-Raf inhibitor PLX4032 induces DR5 upregulation exclusively in Ras-mutant cancer cells; this effect is dependent on Ras/c-Raf/MEK/ERK signaling activation. PLX4032 induces DR5 expression at transcriptional levels, largely due to enhancing CHOP/Elk1-mediated DR5 transcription. Pre-exposure of Ras-mutated cancer cells to PLX4032 sensitizes them to TRAIL-induced apoptosis; this is also a c-Raf/MEK/ERK-dependent event. Collectively, our findings highlight a previously undiscovered effect of B-Raf inhibition on the induction of DR5 expression and the enhancement of DR5 activation-induced apoptosis in Ras-mutant cancer cells and hence may suggest a novel therapeutic strategy against Ras-mutated cancer cells by driving their death due to DR5-dependent apoptosis through B-Raf inhibition.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes ras , Indóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Sulfonamidas/farmacologia , Apoptose/efeitos dos fármacos , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Proteínas de Neoplasias/genética , Oximas/farmacologia , Proteínas Proto-Oncogênicas B-raf/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteínas Recombinantes/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Transcrição CHOP/fisiologia , Transcrição Gênica/efeitos dos fármacos , Vemurafenib , Proteínas Elk-1 do Domínio ets/fisiologia
11.
J Transl Med ; 14(1): 106, 2016 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-27118475

RESUMO

BACKGROUND: Chronic lymphocytic leukemia (CLL) remains incurable with standard therapy, and is characterized by excessive expansion of monoclonal abnormal mature B cells and more regulatory immune properties of T cell compartment. Thus, developing novel strategies to enhance immune function merits further investigation as a possible therapy for CLL. METHODS: We generated a fusion cytokine (fusokine) arising from the combination of human GM-CSF and IL-4 (named GIFT4). Primary CLL cells were treated with GIFT4 or GM-CSG and IL-4 in vitro. GIFT4-triggered STAT5 signaling in CLL cells was examined by Western blot. The phenotype and secretome of GIFT4-treated CLL cells (GIFT4-CLL cells), and the immune stimulatory function of GIFT4-CLL cells on autologous T cells were analyzed by flow cytometry and luminex assay. RESULTS: GIFT4-CLL up-regulated the expression of co-stimulatory molecules CD40, CD80 and CD86 and adhesion molecule CD54. GIFT4-CLL cells secreted IL-1ß, IL-6, ICAM-1 and substantial IL-2 relative to unstimulated CLL cells. GIFT4 treatment led to JAK1, JAK2 and JAK3-mediated hyper-phosphorylation of STAT5 in primary CLL cells, which is essential for GIFT4-triggered conversion of CLL cells. GIFT4-CLL cells directly propelled the expansion of autologous IFN-γ-producing CD314(+) cytotoxic T cells in vitro, and that these could lyse autologous CLL cells. Furthermore, administration of GIFT4 protein promoted the expansion of human T cells in NOD-scid IL2Rγ(null) immune deficient mice adoptively pre-transferred with peripheral blood mononuclear cells from subjects with CLL. CONCLUSION: GIFT4 has potent capability to converts primary CLL cells into APC-like immune helper cells that initiate a T cell driven anti-CLL immune response.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-4/farmacologia , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/patologia , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T Auxiliares-Indutores/imunologia , Adulto , Idoso , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/metabolismo , Proliferação de Células/efeitos dos fármacos , Apresentação Cruzada/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Feminino , Humanos , Janus Quinases/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Fenótipo , Proteoma/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos
12.
Oncotarget ; 7(19): 27753-63, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27049723

RESUMO

Multiple myeloma (MM) is a heterogeneous plasma cell malignancy and remains incurable. B-cell lymphoma-2 (BCL2) protein correlates with the survival and the drug resistance of myeloma cells. BH3 mimetics have been developed to disrupt the binding between BCL2 and its pro-apoptotic BCL2 family partners for the treatment of MM, but with limited therapeutic efficacy. We recently identified a small molecule BDA-366 as a BCL2 BH4 domain antagonist, converting it from an anti-apoptotic into a pro-apoptotic molecule. In this study, we demonstrated that BDA-366 induces robust apoptosis in MM cell lines and primary MM cells by inducing BCL2 conformational change. Delivery of BDA-366 substantially suppressed the growth of human MM xenografts in NOD-scid/IL2Rγnull mice, without significant cytotoxic effects on normal hematopoietic cells or body weight. Thus, BDA-366 functions as a novel BH4-based BCL2 inhibitor and offers an entirely new tool for MM therapy.


Assuntos
Antraquinonas/uso terapêutico , Apoptose/efeitos dos fármacos , Etanolaminas/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Animais , Antraquinonas/efeitos adversos , Linhagem Celular Tumoral , Etanolaminas/efeitos adversos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-bcl-2/química , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Sci Rep ; 5: 11856, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26150163

RESUMO

HIV vaccines should elicit immune responses at both the mucosal portals of entry to block transmission and systemic compartments to clear disseminated viruses. Co-delivery of mucosal adjuvants has been shown to be essential to induce effective mucosal immunity by non-replicating vaccines. A novel cytokine, GIFT4, engineered by fusing GM-CSF and interleukin-4, was previously found to simulate B cell proliferation and effector function. Herein a membrane-anchored form of GIFT4 was constructed by fusing a glycolipid (GPI)-anchoring sequence and incorporated into Env-enriched HIV virus-like particles (VLPs) as a molecular adjuvant. Guinea pigs were immunized with the resulting HIV VLPs through an intramuscular priming-intranasal boosting immunization route. The GIFT4-containing VLPs induced higher levels of systemic antibody responses with significantly increased binding avidity and improved neutralizing breadth and potency to a panel of selected strains, as well as higher levels of IgG and IgA at several mucosal sites. Thus, the novel GPI-GIFT4-containging VLPs have the potential to be developed into a prophylactic HIV vaccine. Incorporation of GPI-anchored GIFT4 into VLPs as a molecular adjuvant represents a novel approach to increase their immunogenicity.


Assuntos
Glicolipídeos/química , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , HIV/imunologia , Interleucina-4/metabolismo , Vacinas de Partículas Semelhantes a Vírus/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos/imunologia , Anticorpos/metabolismo , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/metabolismo , Afinidade de Anticorpos/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Cobaias , Humanos , Imunidade nas Mucosas , Imunoglobulina A/sangue , Imunoglobulina A/metabolismo , Imunoglobulina G/sangue , Imunoglobulina G/metabolismo , Interleucina-4/genética , Vacinas de Partículas Semelhantes a Vírus/genética , Vacinas de Partículas Semelhantes a Vírus/metabolismo
14.
Oncotarget ; 6(19): 17532-42, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26009898

RESUMO

Carfilzomib (CFZ) is a second generation proteasome inhibitor approved for the treatment of patients with multiple myeloma. It induces apoptosis in human cancer cells; but the underlying mechanisms remain undefined. In the present study, we show that CFZ decreases the survival of several human cancer cell lines and induces apoptosis. Induction of apoptosis by CFZ occurs, at least in part, due to activation of the extrinsic apoptotic pathway, since FADD deficiency protected cancer cells from undergoing apoptosis. CFZ increased total and cell surface levels of DR5 in different cancer cell lines; accordingly it enhanced TRAIL-induced apoptosis. DR5 deficiency protected cancer cells from induction of apoptosis by CFZ either alone or in combination with TRAIL. These data together convincingly demonstrate that DR5 upregulation is a critical mechanism accounting for CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis. CFZ inhibited the degradation of DR5, suggesting that DR5 stabilization contributes to CFZ-induced DR5 upregulation. In summary, the present study highlights the important role of DR5 upregulation in CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis in human cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Oligopeptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Western Blotting , Linhagem Celular Tumoral , Humanos , Reação em Cadeia da Polimerase , RNA Interferente Pequeno , Transfecção , Regulação para Cima
15.
Exp Hematol Oncol ; 3(1): 27, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25422792

RESUMO

OBJECTIVES: Multiple myeloma (MM) is an incurable B-cell cancer with accumulated clonal abnormal plasma cells in bone marrow of patients. MCL-1 (myeloid cell leukemia sequence 1) protein is an anti-apoptotic molecule in MM cells and regulated by pro-inflammatory cytokine IL-6 and downstream signaling molecules STAT3, PI3K and MAPK. The purpose of this study is to investigate the effect of STAT3, PI3K and MAPK gene silence on MCL-1 expression in human MM cells and the consequence of cell survival. METHODS: Lentivirus small hairpin RNA (shRNA) interference techniques were utilized to knock down STAT3, PI3K or MAPK genes. Gene and protein expression was quantified by quantitative real-time PCR and Western Blot. MM cell apoptosis was examined by annexin-V FITC/propidium iodide staining. RESULTS: Efficient silence of STAT3, PI3K, MAPK1 or MAPK2 gene robustly abrogated IL-6 enhanced MCL-1 expression and suppressed MM cell growth. Silencing STAT3 gene inhibited PI3K expression, silencing PI3K markedly abrogated STAT3 and MAPK production. Inhibition of MAPK2 gene by shMAPK2 suppressed STAT3, PI3K and MAPK1 expression in the cells. Silencing of STAT3, PI3K and MAPK2 together completely blocked MCL-1 expression in MM cells. CONCLUSION: There is a syngeneic effect among the three independent STAT3, PI3K and MAPK2 survival-signaling pathways related to MCL-1 expression in MM cells. shRNAs silencing of STAT3, PI3K and MAPK2 together could provide an effective strategy to treat MM.

16.
PLoS One ; 9(9): e106724, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25198691

RESUMO

Fusokines are chimeric proteins generated by the physical coupling of cytokines in a single polypeptide, resulting in proteins with highly pleiotropic activity and the potential to treat cancer and autoimmune ailments. For instance, the fusokine GIFT15 (GM-CSF and Interleukin 15 Fusion Transgene) has been shown to be a powerful immunosuppressive protein able to convert naïve B cells into IL-10-producing B cells. To date, the mammalian cell systems used for the expression of GIFT15 allow for secretion of the protein in the culturing media, an inefficient system for producing GMP-compliant fusokines. In this study we report the bacterial expression of bioactive recombinant GIFT15 (rGIFT15). Indeed, there is a constant demand to improve the expression systems for therapeutic proteins. Expression of a maltose-binding protein (MBP) fusion protein efficiently allowed the accumulation of soluble protein in the intracellular milieu. Optimizing the bacterial culture significantly increased the yield of recombinant protein. The biological activity of rGIFT15 was comparable to that of fusokine derived from a mammalian source. This approach led to the production of soluble, endotoxin-free functional protein, averaging 5 mg of rGIFT15 per liter of culture. This process is amenable to scale up for the development of Food and Drug Administration (FDA)-compliant immune-modulatory rGIFT15.


Assuntos
Bactérias/metabolismo , Citocinas/metabolismo , Proteínas Ligantes de Maltose/genética , Proteínas Recombinantes de Fusão/genética , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Camundongos
17.
Methods Mol Biol ; 1190: 115-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25015277

RESUMO

Naïve B cells are crucial components of adaptive immunity. In addition to their capacity to produce immunoglobulins, a minor subset termed regulatory B cells or Bregs has been proven to modulate inflammation through the secretion of soluble mediators. The two main technical difficulties with their clinical use lie in their relatively low abundance in vivo and the scarcity of known methods for their ex vivo expansion. While studying the pharmacological properties of a novel bifunctional granulocyte macrophage-colony-stimulating factor (GM-CSF) and IL-15 fusion transgene (GIFT15) on unfractionated splenocytes in vitro, we observed that the GIFT15 fusokine had a remarkable and unprecedented effect on naïve B cells by converting them into suppressor cells of B-cell ontogeny (hereafter referred to as GIFT15 inducible (i)Bregs). Moreover, GIFT15 promoted iBreg proliferation. We present in this report a detailed protocol using the GIFT15 fusokine as a tool for the ex vivo generation of murine iBregs, which may serve as an immediate remedy to their abundance challenge in the clinic.


Assuntos
Linfócitos B/efeitos dos fármacos , Citocinas/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-10/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B Reguladores/citologia , Linfócitos B Reguladores/efeitos dos fármacos , Linfócitos B Reguladores/imunologia , Separação Celular/métodos , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia
18.
Cancer Res ; 74(15): 4133-44, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24938765

RESUMO

Engineered chimeric cytokines can generate gain-of-function activity in immune cells. Here, we report potent antitumor activity for a novel fusion cytokine generated by N-terminal coupling of GM-CSF to IL4, generating a fusokine termed GIFT4. B cells treated with GIFT4 clustered GM-CSF and IL4 receptors on the cell surface and displayed a pan-STAT hyperphosphorylation associated with acquisition of a distinct phenotype and function described to date. In C57BL/6J mice, administration of GIFT4 expanded endogenous B cells and suppressed the growth of B16F0 melanoma cells. Furthermore, B16F0 melanoma cells engineered to secrete GIFT4 were rejected immunologically in a B-cell-dependent manner. This effect was abolished when GIFT4-expressing B16F0 cells were implanted in B-cell-deficient mice, confirming a B-cell-dependent antitumor effect. Human GIFT4-licensed B cells primed cytotoxic T cells and specifically killed melanoma cells in vitro and in vivo. Taken together, our results demonstrated that GIFT4 could mediate expansion of B cells with potent antigen-specific effector function. GIFT4 may offer a novel immunotherapeutic tool and define a previously unrecognized potential for B cells in melanoma immunotherapy.


Assuntos
Linfócitos B/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Interleucina-4/farmacologia , Melanoma Experimental/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T Citotóxicos/imunologia , Animais , Processos de Crescimento Celular/imunologia , Engenharia Genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Interleucina-4/genética , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Engenharia de Proteínas/métodos , Proteínas Recombinantes de Fusão/genética
19.
Oncoimmunology ; 3(10): e955702, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25941574

RESUMO

We have engineered a novel fusion cytokine named GIFT4 derived from GM-CSF and IL-4, and displaying robust gain-of-function immunostimulatory effects on B cells. GIFT4-programmed B cells have a unique identity and potent capacity to elicit a tumoricidal - cell response, thus comprising a novel B cell-based cancer immunotherapeutic approach.

20.
Infect Disord Drug Targets ; 12(3): 248-54, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22394176

RESUMO

B cells play a pivotal role in host adaptive immunity against pathogenic microorganisms, but may also maladaptively contribute to the pathogenesis of autoimmune diseases. In contrast, distinct B cell subsets have the capacity to regulate host immune response, and suppress inflammation. B regulatory cells are a rare population of endogenous Blymphocytes defined in part by production of the anti-inflammatory cytokine IL-10. Although "natural" B regulatory cells exist in vivo, the low frequency of B regulatory cells may be a limiting factor on their impact in autoimmune ailments. In answer to this unmet need, we have developed a novel strategy for alternate lymphoid activation: fusokines. These wholly engineered chimeric leukines fuse two functionally unrelated cytokines for the purpose of alternate immune modulation. The GM-CSF- and IL-15-derived fusokine: GIFT15, possesses entirely novel and unheralded immune modulating properties mediated through the IL15 receptor which reprograms naive B cells into B regulatory cells (Bregs). In this article, we review the current approaches to generate Bregs in vitro, and highlight gain-of-function mechanisms by which GIFT15- induced Bregs abrogate pathogenic autoimmunity in mice. We also demonstrate that the human equivalent of inducible Bregs may also serve as a new potent therapeutic tool for treatment of autoimmune disease.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B Reguladores/imunologia , Citocinas/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Autoimunidade/imunologia , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interleucina-10/imunologia , Interleucina-15/imunologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA