Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Signal Transduct Target Ther ; 8(1): 456, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38105247

RESUMO

Metastasis is the leading cause of cancer-related deaths. Transforming growth factor beta (TGF-ß) signaling drives metastasis and is strongly enhanced during cancer progression. Yet, the use of on-target TGF-ß signaling inhibitors in the treatment of cancer patients remains unsuccessful, highlighting a gap in the understanding of TGF-ß biology that limits the establishment of efficient anti-metastatic therapies. Here, we show that TGF-ß signaling hyperactivation in breast cancer cells is required for metastasis and relies on increased small extracellular vesicle (sEV) secretion. Demonstrating sEV's unique role, TGF-ß signaling levels induced by sEVs exceed the activity of matching concentrations of soluble ligand TGF-ß. Further, genetic disruption of sEV secretion in highly-metastatic breast cancer cells impairs cancer cell aggressiveness by reducing TGF-ß signaling to nearly-normal levels. Otherwise, TGF-ß signaling activity in non-invasive breast cancer cells is inherently low, but can be amplified by sEVs, enabling invasion and metastasis of poorly-metastatic breast cancer cells. Underscoring the translational potential of inhibiting sEV trafficking in advanced breast cancers, treatment with dimethyl amiloride (DMA) decreases sEV secretion, TGF-ß signaling activity, and breast cancer progression in vivo. Targeting both the sEV trafficking and TGF-ß signaling by combining DMA and SB431542 at suboptimal doses potentiated this effect, normalizing the TGF-ß signaling in primary tumors to potently reduce circulating tumor cells, metastasis, and tumor self-seeding. Collectively, this study establishes sEVs as critical elements in TGF-ß biology, demonstrating the feasibility of inhibiting sEV trafficking as a new therapeutic approach to impair metastasis by normalizing TGF-ß signaling levels in breast cancer cells.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , Humanos , Feminino , Linhagem Celular Tumoral , Fator de Crescimento Transformador beta/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo
2.
Am J Pathol ; 193(8): 1029-1045, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37236504

RESUMO

IL-33, a member of the IL-1 family, acts as an alarmin in immune response. Epithelial-mesenchymal transition and transforming growth factor-ß (TGF-ß)­induced fibroblast activation are key events in the development of renal interstitial fibrosis. The current study found increased expression of IL-33 and interleukin-1 receptor-like 1 (IL1RL1, alias ST2), the receptor for IL-33, in human fibrotic renal tissues. In addition, IL-33­ or ST2-deficient mice showed significantly reduced levels of fibronectin, α-smooth muscle actin, and vimentin, and increased E-cadherin levels. In HK-2 cells, IL-33 promotes the phosphorylation of the TGF-ß receptor (TGF-ßR), Smad2, and Smad3, and the production of extracellular matrix (ECM), with reduced expression of E-cadherin. Blocking TGF-ßR signaling or suppressing ST2 expression impeded Smad2 and Smad3 phosphorylation, thereby reducing ECM production, suggesting that IL-33­induced ECM synthesis requires cooperation between the two pathways. Mechanistically, IL-33 treatment induced a proximate interaction between ST2 and TGF-ßRs, activating downstream Smad2 and Smad3 for ECM production in renal epithelial cells. Collectively, this study identified a novel and essential role for IL-33 in promoting TGF-ß signaling and ECM production in the development of renal fibrosis. Therefore, targeting IL-33/ST2 signaling may be an effective therapeutic strategy for renal fibrosis.


Assuntos
Interleucina-33 , Nefropatias , Camundongos , Humanos , Animais , Interleucina-33/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Nefropatias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Smad3/metabolismo , Fibrose , Caderinas/metabolismo , Fatores de Crescimento Transformadores/metabolismo , Fatores de Crescimento Transformadores/farmacologia , Fatores de Crescimento Transformadores/uso terapêutico , Fator de Crescimento Transformador beta1/metabolismo , Transição Epitelial-Mesenquimal
3.
BMC Med ; 20(1): 408, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36280870

RESUMO

BACKGROUND: Dual inhibition of PD-1/PD-L1 and TGF-ß pathways is a rational therapeutic strategy for malignancies. SHR-1701 is a new bifunctional fusion protein composed of a monoclonal antibody against PD-L1 fused with the extracellular domain of TGF-ß receptor II. This first-in-human trial aimed to assess SHR-1701 in pretreated advanced solid tumors and find the population who could benefit from SHR-1701. METHODS: This was a dose-escalation, dose-expansion, and clinical-expansion phase 1 study. Dose escalation was initiated by accelerated titration (1 mg/kg q3w; intravenous infusion) and then switched to a 3+3 scheme (3, 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w), followed by dose expansion at 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w. The primary endpoints of the dose-escalation and dose-expansion parts were the maximum tolerated dose and recommended phase 2 dose. In the clinical-expansion part, selected tumors were enrolled to receive SHR-1701 at the recommended dose, with a primary endpoint of confirmed objective response rate (ORR). RESULTS: In total, 171 patients were enrolled (dose-escalation: n=17; dose-expansion, n=33; clinical-expansion, n=121). In the dose-escalation part, no dose-limiting toxicity was observed, and the maximum tolerated dose was not reached. SHR-1701 showed a linear dose-exposure relationship and the highest ORR at 30 mg/kg every 3 weeks, without obviously aggravated toxicities across doses in the dose-escalation and dose-expansion parts. Combined, 30 mg/kg every 3 weeks was determined as the recommended phase 2 dose. In the clinical-expansion part, SHR-1701 showed the most favorable efficacy in the gastric cancer cohort, with an ORR of 20.0% (7/35; 95% CI, 8.4-36.9) and a 12-month overall survival rate of 54.5% (95% CI, 29.5-73.9). Grade ≥3 treatment-related adverse events occurred in 37 of 171 patients (22%), mainly including increased gamma-glutamyltransferase (4%), increased aspartate aminotransferase (3%), anemia (3%), hyponatremia (3%), and rash (2%). Generally, patients with PD-L1 CPS ≥1 or pSMAD2 histochemical score ≥235 had numerically higher ORR. CONCLUSIONS: SHR-1701 showed an acceptable safety profile and encouraging antitumor activity in pretreated advanced solid tumors, especially in gastric cancer, establishing the foundation for further exploration. TRIAL REGISTRATION: ClinicalTrials.gov , NCT03710265.


Assuntos
Neoplasias Gástricas , Humanos , gama-Glutamiltransferase/uso terapêutico , Receptor de Morte Celular Programada 1 , Anticorpos Monoclonais/uso terapêutico , Aspartato Aminotransferases/uso terapêutico , Fator de Crescimento Transformador beta/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico
4.
J Orthop Surg Res ; 13(1): 72, 2018 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-29622010

RESUMO

BACKGROUND: TGF-ß has been described as a mediator of fibrosis and scarring. Several studies achieved reduction in experimental scarring through the inhibition of TGF-ß. Fibroblasts have been defined as the cell population originating fibrosis, blocking fibroblast invasion may impair epidural fibrosis appearance. For this purpose, biocompatible materials used as mechanical barriers and a TGF-ß inhibitor peptide were evaluated in the reduction of epidural fibrosis. METHODS: A L6 laminectomy was performed in 40 New Zealand white rabbits. Divided into four groups, each rabbit was assigned to receive either collagen sponge scaffold (CS group), gelatin-based gel (GCP group), P144® (iTGFß group), or left untreated (control group). Four weeks after surgery, cell density, collagen content, and new bone formation of the scar area were determined by histomorphometry. Two experienced pathologists scored dura mater adhesion, scar density, and inflammatory infiltrate in a blinded manner. RESULTS: In all groups, laminectomy site was filled with fibrous tissue and the dura mater presented adhesions. Only GCP group presented a significant reduction in collagen content and scar density. CONCLUSION: GCP treatment reduces epidural fibrosis although did not prevent dura mater adhesion completely.


Assuntos
Espaço Epidural/patologia , Laminectomia/efeitos adversos , Fragmentos de Peptídeos/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Aderências Teciduais/prevenção & controle , Animais , Materiais Biocompatíveis , Cicatriz/etiologia , Cicatriz/patologia , Cicatriz/prevenção & controle , Colágeno/metabolismo , Modelos Animais de Doenças , Dura-Máter/metabolismo , Dura-Máter/patologia , Espaço Epidural/metabolismo , Fibrose , Masculino , Compostos Orgânicos/uso terapêutico , Coelhos , Aderências Teciduais/etiologia , Aderências Teciduais/patologia , Fator de Crescimento Transformador beta/antagonistas & inibidores
5.
Urol Oncol ; 36(3): 93.e1-93.e11, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29103968

RESUMO

BACKGROUND: Avelumab has recently been approved by the Food and Drug Administration for the therapy of Merkel cell carcinoma and urothelial carcinoma. M7824 is a novel first-in-class bifunctional fusion protein comprising a monoclonal antibody against programmed death-ligand 1 (PD-L1, avelumab), fused to the extracellular domain of human transforming growth factor beta (TGFß) receptor 2, which functions as a TGFß "trap." Advanced urothelial tumors have been shown to express TGFß, which possesses immunosuppressive properties that promote cancer progression and metastasis. The rationale for a combined molecule is to block the PD-1/PD-L1 interaction between tumor cells and immune cell infiltrate and simultaneously reduce or eliminate TGFß from the tumor microenvironment. In this study, we explored the effect of M7824 on invasive urothelial carcinoma cell lines. METHODS: Human urothelial (transitional cell) carcinoma cell lines HTB-4, HTB-1, and HTB-5 were treated with M7824, M7824mut (M7824 that is mutated in the anti-PD-L1 portion of the molecule and thus does not bind PD-L1), anti-PD-L1 (avelumab), or IgG1 isotype control monoclonal antibody, and were assessed for gene expression, cell-surface phenotype, and sensitivity to lysis by TRAIL, antigen-specific cytotoxic T lymphocytes and natural killer cells. RESULTS: M7824 retains the ability to mediate antibody-dependent cellular cytotoxicity of tumor cells, although in some cases to a lesser extent than anti-PD-L1. However, compared to anti-PD-L1, M7824 increases (A) gene expression of molecules involved in T-cell trafficking in the tumor (e.g., CXCL11), (B) TRAIL-mediated tumor cell lysis, and (C) antigen-specific CD8+ T-cell-mediated lysis of tumor cells. CONCLUSIONS: These studies demonstrate the immunomodulatory properties of M7824 on both tumor cell phenotype and immune-mediated lysis. Compared to anti-PD-L1 or M7824mut, M7824 induces immunogenic modulation of urothelial carcinoma cell lines, rendering them more susceptible to immune-mediated recognition and lysis. These findings show the relevance of the dual blockade of PD-L1 and TGFß in urothelial carcinoma cell lines and thus support the rationale for future clinical studies of M7824 in patients with urothelial cancer.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células de Transição/tratamento farmacológico , Proteínas Recombinantes de Fusão/farmacologia , Neoplasias Urológicas/tratamento farmacológico , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Carcinoma de Células de Transição/imunologia , Carcinoma de Células de Transição/patologia , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/farmacologia , Proteínas Serina-Treonina Quinases/uso terapêutico , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/uso terapêutico , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Neoplasias Urológicas/imunologia , Neoplasias Urológicas/patologia
6.
Anticancer Res ; 34(12): 6939-44, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25503119

RESUMO

ß-glucans are well-established immunomodulators with strong effects resulting in slowing or even inhibiting cancer growth. Recent studies have repeatedly suggested that the biological activities of ß-glucan can be potentiated by the addition of other bioactive agents. In the current study, we focused on the anticancer effects of a combination of yeast-derived ß-glucan and a selenium-linked pseudodisaccharide. Using three different models of murine cancer, we showed that this combination strongly suppressed the growth of all three types of cancers, most likely via the interaction with natural anticancer antibodies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Glucanos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Compostos Organosselênicos/uso terapêutico , Proteoglicanas/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Linhagem Celular Tumoral , Ciclofosfamida/uso terapêutico , Dissacarídeos/uso terapêutico , Feminino , Humanos , Fatores Imunológicos/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Resveratrol , Estilbenos/uso terapêutico
7.
Antioxid Redox Signal ; 19(14): 1607-18, 2013 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-23350688

RESUMO

AIMS: The NADPH oxidases constitute a major source of superoxide anion (·O2(-)) in hypertension. Several studies suggest an important role of NADPH oxidases in different effects mediated by transforming growth factor-ß1 (TGF-ß1). We investigated whether a chronic treatment with P144, a peptide synthesized from type III TGF-ß1 receptor, inhibited NADPH oxidases in the renal cortex of spontaneously hypertensive rats (SHR). RESULTS: Here, we show that chronic administration of P144 significantly reduced the NADPH oxidase expression and activity as well as the oxidative stress observed in control vehicle-treated SHR (V-SHR). In addition, P144 was also able to reduce the significant increase in the renal fibrosis and in mRNA expression of different components of collagen metabolism, as well as in the levels of connective tissue growth factor observed in V-SHR. Finally, TGF-ß1-stimulated NRK52E exhibited a significant increase in NADPH oxidase expression and activity as well as a TGF-ß1-dependent intracellular pathway that were inhibited in the presence of P144. INNOVATION: Our experimental evidence suggests that reversing oxidative stress may be therapeutically useful in preventing fibrosis-associated renal damage. We show here that (i) the TGF-ß1-NADPH oxidases axis is crucial in the development of fibrosis in an experimental hypertensive renal disease animal model, and (ii) the use of P144 reverses TGF-ß1-dependent NADPH oxidase activity; thus, P144 may be considered a novel therapeutic tool in kidney disease associated with hypertension. CONCLUSION: We demonstrate that P144 inhibits NADPH oxidases and prevents oxidative stress in kidneys from hypertensive rats. Our data also suggest that these effects are associated with the renal antifibrotic effect of P144.


Assuntos
Rim/efeitos dos fármacos , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Proteoglicanas/química , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Animais , Fibrose/metabolismo , Fibrose/prevenção & controle , Rim/metabolismo , Rim/patologia , Masculino , Ratos , Ratos Endogâmicos SHR , Transdução de Sinais/efeitos dos fármacos
8.
Exp Cell Res ; 319(3): 12-22, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23153552

RESUMO

Colorectal cancer (CRC) frequently metastasizes to the liver, a phenomenon that involves the participation of transforming-growth-factor-ß(1) (TGFß(1)). Blockade of the protumorigenic effects elicited by TGFß(1) in advanced CRC could attenuate liver metastasis. We aimed in the present study to assess the antimetastatic effect of TGFß(1)-blocking peptides P17 and P144, and to study mechanisms responsible for this activity in a mouse model. Colon adenocarcinoma cells expressing luciferase were pretreated with TGFß(1) (Mc38-luc(TGFß1) cells), injected into the spleen of mice and monitored for tumor development. TGFß(1) increased primary tumor growth and liver metastasis, whereas systemic treatment of mice with either P17 or P144 significantly reduced tumor burden (p<0.01). In metastatic nodules, mitotic/apoptotic ratio, mesenchymal traits and angiogenesis (evaluated by CD-31, as well as circulating endothelial and progenitor cells) induced by TGFß(1) were consistently reduced following injection of peptides. In vitro experiments revealed a direct effect of TGFß(1) in Mc38 cells, which resulted in activation of Smad2, Smad3 and Smad1/5/8, and increased invasion and transendothelial migration, whereas blockade of TGFß(1)-signaling reverted these features. Because TGFß(1)-mediated epithelial-mesenchymal transition (EMT) has been suggested to induce a cancer stem cell (CSC) phenotype, we analyzed the ability of this cytokine to induce tumorsphere formation and the expression of CSC markers. In TGFß(1)-treated cells, tumorspheres were enriched in CD44 and SOX2, which were diminished in the presence of P17. Our data provide a preclinical rationale to evaluate P17 and P144 as potential therapeutic options for the treatment of metastatic CRC.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias do Colo/tratamento farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Hepáticas/prevenção & controle , Células-Tronco Neoplásicas/efeitos dos fármacos , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Adenocarcinoma/patologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Células Cultivadas , Neoplasias do Colo/patologia , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Hepáticas/secundário , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/patologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Peptídeos/administração & dosagem , Peptídeos/farmacologia , Fenótipo , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Fator de Crescimento Transformador beta1/antagonistas & inibidores
9.
Oxid Med Cell Longev ; 2012: 726940, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701756

RESUMO

NADPH oxidases constitute a major source of superoxide anion (·O(2)(-)) in hypertension. Several studies suggest an important role of NADPH oxidases in different effects mediated by TGF-ß 1. In this study we show that chronic administration of P144, a peptide synthesized from type III TGF-ß 1 receptor, significantly reduced the cardiac NADPH oxidase expression and activity as well as in the nitrotyrosine levels observed in control spontaneously hypertensive rats (V-SHR) to levels similar to control normotensive Wistar Kyoto rats. In addition, P144 was also able to reduce the significant increases in the expression of collagen type I protein and mRNA observed in hearts from V-SHR. In addition, positive correlations between collagen expression, NADPH oxidase activity, and nitrotyrosine levels were found in all animals. Finally, TGF-ß 1-stimulated Rat-2 exhibited significant increases in NADPH oxidase activity that was inhibited in the presence of P144. It could be concluded that the blockade of TGF-ß 1 with P144 inhibited cardiac NADPH oxidase in SHR, thus adding new data to elucidate the involvement of this enzyme in the profibrotic actions of TGF-ß 1.


Assuntos
Hipertensão/enzimologia , NADPH Oxidases/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Pressão Sanguínea/efeitos dos fármacos , Linhagem Celular , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Fibroblastos/efeitos dos fármacos , Coração/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Miocárdio/metabolismo , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genética , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Ratos , Ratos Endogâmicos WKY , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Fator de Crescimento Transformador beta1/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
10.
J Am Soc Nephrol ; 22(9): 1682-95, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21742730

RESUMO

During peritoneal dialysis (PD), mesothelial cells undergo mesothelial-to-mesenchymal transition (MMT), a process associated with peritoneal-membrane dysfunction. Because TGF-ß1 can induce MMT, we evaluated the efficacy of TGF-ß1-blocking peptides in modulating MMT and ameliorating peritoneal damage in a mouse model of PD. Exposure of the peritoneum to PD fluid induced fibrosis, angiogenesis, functional impairment, and the accumulation of fibroblasts. In addition to expressing fibroblast-specific protein-1 (FSP-1), some fibroblasts co-expressed cytokeratin, indicating their mesothelial origin. These intermediate-phenotype (Cyto(+)/FSP-1(+)) fibroblasts had features of myofibroblasts with fibrogenic capacity. PD fluid treatment triggered the appearance of CD31(+)/FSP-1(+) and CD45(+)/FSP-1(+) cells, suggesting that fibroblasts also originate from endothelial cells and from cells recruited from bone marrow. Administration of blocking peptides significantly ameliorated fibrosis and angiogenesis, improved peritoneal function, and reduced the number of FSP-1(+) cells, especially in the Cyto(+)/FSP-1(+) subpopulation. Conversely, overexpression of TGF-ß1 in the peritoneum by adenovirus-mediated gene transfer led to a marked accumulation of fibroblasts, most of which derived from the mesothelium. Taken together, these results demonstrate that TGF-ß1 drives the peritoneal deterioration induced by dialysis fluid and highlights a role of TGF-ß1-mediated MMT in the pathophysiology of peritoneal-membrane dysfunction.


Assuntos
Transdiferenciação Celular/efeitos dos fármacos , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , Peritônio/patologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Biomarcadores/metabolismo , Células Cultivadas , Soluções para Diálise/efeitos adversos , Feminino , Injeções Intraperitoneais , Queratinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Fibrose Peritoneal/prevenção & controle , Fenótipo , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores
11.
Int J Radiat Oncol Biol Phys ; 78(5): 1513-23, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20932668

RESUMO

PURPOSE: We determined whether anti-transforming growth factor-ß (TGF-ß) intervention could halt the progression of established radiation-induced liver fibrosis (RILF). METHODS AND MATERIALS: A replication-defective adenoviral vector expressing the extracellular portion of human TßRII and the Fc portion of immunoglobulin G fusion protein (AdTßRIIFc) was produced. The entire rat liver was exposed to 30 Gy irradiation to generate a RILF model (RILFM). Then, RILFM animals were treated with AdTßRIIFc (1 × 10(11) plaque-forming units [PFU] of TßRII), control virus (1 × 10(11) PFU of AdGFP), or saline. Delayed radiation liver injury was assessed by histology and immunohistochemistry. Chronic oxidative stress damage, hepatic stellate cell activation, and hepatocyte regeneration were also analyzed. RESULTS: In rats infected with AdTßRIIFc, fibrosis was significantly improved compared with rats treated with AdGFP or saline, as assessed by histology, hydroxyproline content, and serum level of hyaluronic acid. Compared with AdGFP rats, AdTßRIIFc-treated rats exhibited decreased oxidative stress damage and hepatic stellate cell activation and preserved liver function. CONCLUSIONS: Our results demonstrate that TGF-ß plays a critical role in the progression of liver fibrosis and suggest that anti-TGF-ß intervention is feasible and ameliorates established liver fibrosis. In addition, chronic oxidative stress may be involved in the progression of RILF.


Assuntos
Terapia Genética/métodos , Cirrose Hepática Experimental/prevenção & controle , Fígado/efeitos da radiação , Lesões Experimentais por Radiação/prevenção & controle , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adenoviridae/genética , Animais , Vetores Genéticos/uso terapêutico , Células Estreladas do Fígado , Ácido Hialurônico/sangue , Hidroxiprolina/análise , Fígado/metabolismo , Cirrose Hepática Experimental/metabolismo , Masculino , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/sangue , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/uso terapêutico , Lesões Experimentais por Radiação/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de IgG/metabolismo , Receptores de IgG/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/sangue , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Fator de Crescimento Transformador beta/metabolismo , Ensaio de Placa Viral/métodos
12.
Cancer Res ; 70(20): 8159-68, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20889724

RESUMO

As a tumor marker for colorectal cancers, carcinoembryonic antigen (CEA) enhances the metastatic potential of cancer cells. CEA functions as an intercellular adhesion molecule and is upregulated in a wide variety of human cancers. However, the molecular mechanisms by which CEA mediates metastasis remain to be understood. Transforming growth factor-ß (TGF-ß) signaling regulates both tumor suppression and metastasis, and also contributes to the stimulation of CEA transcription and secretion in colorectal cancer cells. However, it remains unknown whether CEA, in turn, influences TGF-ß functions and if a regulatory cross-talk exists between CEA and the TGF-ß signaling pathway. Here, we report that CEA directly interacts with TGF-ß receptor and inhibits TGF-ß signaling. Targeting CEA with either CEA-specific antibody or siRNA rescues TGF-ß response in colorectal cancer cell lines with elevated CEA, thereby restoring the inhibitory effects of TGF-ß signaling on proliferation. CEA also enhances the survival of colorectal cancer cells in both local colonization and liver metastasis in animal study. Our study provides novel insights into the interaction between CEA and TGF-ß signaling pathway and establishes a negative feedback loop in amplifying the progression of colon cancer cells to more invasive phenotypes. These findings offer new therapeutic opportunities to inhibit colorectal cancer cell proliferation by cotargeting CEA in promoting tumor-inhibitory action of the TGF-ß pathway.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Antígeno Carcinoembrionário/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Divisão Celular , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Neoplasias Colorretais/fisiopatologia , Primers do DNA , Genes myc , Humanos , Repetições de Microssatélites/fisiologia , Microscopia Confocal , Metástase Neoplásica , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia
13.
Hum Gene Ther ; 21(11): 1623-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20712434

RESUMO

We have investigated whether systemic delivery of an oncolytic adenovirus, Ad.sTßRFc, expressing the soluble form of transforming growth factor-ß receptor II fused with human immunoglobulin Fc fragment (sTGFßRIIFc), could inhibit breast cancer bone metastasis in a mouse model. MDA-MB-231 (human breast cancer) cells were inoculated into the left heart ventricles of nude mice. Once the skeletal tumors were visible by X-rays, mice were intravenously injected with either buffer, Ad.sTßRFc, Ad(E1⁻).sTßRFc (a replication-deficient adenovirus expressing sTGFßRIIFc), or Ad.luc2 (a replicating adenovirus expressing firefly luciferase gene). On days 2 and 7 after viral injections, viral replication and sTGFßRIIFc expression were detected in the skeletal tumors in Ad.sTßRFc-treated group; only viral replication in Ad.luc2 group, and sTGFßRIIFc expression in the Ad(E1⁻).sTßRFc group, were detected. To examine the therapeutic effects, buffer or various viral vectors were administered on days 4 and 7 after intracardiac injection of MDA-MB-231 cells. On day 28, X-ray radiography showed a highly significant reduction in lesion size by Ad.sTßRFc, a significant reduction by Ad.luc2, and some reduction by Ad(E1⁻).sTßRFc. Goldner's trichrome and hematoxylin-eosin staining of the bone sections revealed a significant reduction of tumor burden in the Ad.sTßRFc group, but not in the Ad(E1⁻).sTßRFc or Ad.luc2 group. There were significant reductions in free calcium levels by Ad.sTßRFc, Ad(E1⁻).sTßRFc, and Ad.luc2; however, only in the Ad.sTßRFc group were calcium levels reduced to the normal values. These results suggest that concomitant viral replication and sTGFßRIIFc production are important to inhibit bone metastasis and osteolysis, and that Ad.sTßRFc could be developed for targeting breast cancer bone metastases.


Assuntos
Adenoviridae/genética , Neoplasias Ósseas/terapia , Neoplasias da Mama/patologia , Proteínas de Fusão Oncogênica/metabolismo , Terapia Viral Oncolítica , Proteínas Serina-Treonina Quinases/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Adenoviridae/fisiologia , Animais , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Terapia Genética , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Camundongos , Camundongos Nus , Vírus Oncolíticos/genética , Vírus Oncolíticos/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Replicação Viral
14.
Eur J Med Res ; 15(4): 152-61, 2010 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-20554496

RESUMO

OBJECTIVE: To examine the in vivo anti-fibrotic effect of rat soluble transforming growth factor beta receptor II (RsTbetaRII) and IFN-gamma fusion protein (RsTbRII-IFN-gamma) in rat hepatic fibrosis model. METHODS: Model rats were divided into five groups and treated i.m. for 8 weeks: 1) fibrotic model group (each rat, 100 microl of 0.9% NaCl day superset-1); 2) RsTbetaRII-IFN-gamma treatment group (each rat, 0.136 mg x day(-1); 3) IFN-gamma treatment group (each rat, 7.5 MU x day(-1); 4) RsTbetaRII treatment group (each rat, 0.048 mg x day(-1); and 5) mixture of IFN-gamma and RsTbetaRII treatment group (each rat, IFN-gamma 7.5 MU x day(-1)+ RsTbetaRII 0.048 mg x day(-1). After treatment, hepatic fibrogenesis was evaluated by histopathological analysis and measurement of collagen III, alpha-smooth muscle actin (alpha-SMA), TGF-beta1, TGF-betaRII and their mRNA. RESULTS: Immunohistochemistry, Western blot and real-time RT-PCR showed that RsTbetaRII-IFN-gamma treatment significantly inhibited liver expression of collagen III, alpha-SMA, TGF-beta1 and TGF-betaRII at both protein and mRNA levels. Histopathological analysis also showed that the enhanced anti-fibrotic effects were achieved in model rats treated with RsTbetaRII-IFN-gamma. CONCLUSION: Our results confirmed that RsTbetaRII-IFN-gamma has the enhanced effects in reversing hepatic fibrosis.


Assuntos
Interferon gama/uso terapêutico , Cirrose Hepática/tratamento farmacológico , Proteínas Serina-Treonina Quinases/uso terapêutico , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Tetracloreto de Carbono/toxicidade , Primers do DNA , Modelos Animais de Doenças , Amplificação de Genes , Interferon gama/genética , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética
15.
J Immunol ; 184(9): 5130-40, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20348421

RESUMO

Patients receiving hematopoietic stem cell transplantation or bone marrow transplantation (BMT) as therapy for various malignancies or autoimmune diseases have an increased risk for infectious complications posttransplant, especially in the lung. We have used BMT in mice and murine gammaherpesvirus, gammaHV-68, to study the efficacy of adaptive immune responses post-BMT. Five weeks posttransplant, mice have fully reconstituted their hematopoietic lineages in both the lung and periphery. When challenged with virus, however, BMT mice have a reduced ability to clear lytic virus from the lung. Defective viral control in BMT mice is not related to impaired leukocyte recruitment or defective APC function. Rather, BMT mice are characterized by defective CD4 cell proliferation, skewing of effector CD4 T cells from a Th1 to a Th17 phenotype, and an immunosuppressive lung environment at the time of infection that includes overexpression of TGF-beta1 and PGE(2) and increased numbers of regulatory T cells. Neither indomethacin treatment to block PG synthesis nor anti-CD25 depletion of regulatory T cells improved antiviral host defense post-BMT. Transplanting mice with transgenic bone marrow expressing a dominant-negative TGF-betaRII under the permissive CD4 promoter created mice in which effector CD4 and CD8 cells were unresponsive to TGF-beta1. Mice with TGF-beta1-nonresponsive effector T cells had restored antiviral immunity and improved Th1 responses post-BMT. Thus, our results indicate that overexpression of TGF-beta1 following myeloablative conditioning post-BMT results in impaired effector T cell responses to viral infection.


Assuntos
Transplante de Medula Óssea/imunologia , Infecções por Herpesviridae/imunologia , Pneumonia Viral/imunologia , Rhadinovirus/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1/efeitos adversos , Fator de Crescimento Transformador beta1/biossíntese , Infecções Tumorais por Vírus/imunologia , Animais , Transplante de Medula Óssea/efeitos adversos , Diferenciação Celular/imunologia , Proliferação de Células , Inibidores do Crescimento/efeitos adversos , Inibidores do Crescimento/biossíntese , Inibidores do Crescimento/genética , Infecções por Herpesviridae/terapia , Infecções por Herpesviridae/virologia , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pneumonia Viral/terapia , Pneumonia Viral/virologia , Proteínas Serina-Treonina Quinases/administração & dosagem , Proteínas Serina-Treonina Quinases/uso terapêutico , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/virologia , Células Th1/imunologia , Células Th1/patologia , Células Th1/virologia , Fator de Crescimento Transformador beta1/genética , Condicionamento Pré-Transplante/métodos , Infecções Tumorais por Vírus/terapia , Infecções Tumorais por Vírus/virologia
16.
Nutr Res ; 29(4): 248-54, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19410976

RESUMO

Obesity-induced insulin resistance has been suggested to be a systemic inflammatory condition with activation of the innate immune system. Animal studies indicate that certain dietary fibers such as (1,3)(1,6)-beta-D-glycans (BDG) have potent effects on immune activity such as increasing the antiinflammatory cytokine interleukin-10 (IL-10) and reducing the secretion of inflammatory factors. Therefore, we hypothesized that BDG consumption improves inflammatory markers and insulin sensitivity in overweight and obese subjects with moderately increased levels of C-reactive protein, indicating subclinical inflammation. We screened 180 overweight and obese subjects for moderately increased C-reactive protein levels on 2 or more occasions, in the absence of any signs of acute infection. Twelve of the subjects met all inclusion criteria and were investigated in a randomized, double-blind, placebo-controlled, crossover design for 2 x 4 weeks (washout > or =4 weeks). Subjects ingested capsules containing 3 x 0.5 g of highly purified BDG or 3 x 0.5 g of placebo (waxy maize starch) daily. Maintenance of the normal diet of the participants and the correct intake of the capsules were monitored, using 6 x 3-day food recording and counting of the provided capsules. Predefined outcome measures were BDG-induced changes in pro and antiinflammatory markers in circulating blood and gene expression in adipose tissue and peripheral insulin sensitivity expressed as M value. The BDG consumption for 4 weeks significantly increased both circulating levels and adipose tissue messenger RNA (mRNA) expression of the antiinflammatory cytokine IL-10 in overweight and obese humans. Insulin sensitivity as well as circulating levels and mRNA expression of proinflammatory cytokines were unaffected by BDG treatment. Increased IL-10 after BDG consumption might be a contributing factor to the known beneficial effects of dietary fiber intake.


Assuntos
Fibras na Dieta/farmacologia , Inflamação/metabolismo , Resistência à Insulina , Interleucina-10/metabolismo , Obesidade/metabolismo , Sobrepeso/metabolismo , Proteoglicanas/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Fibras na Dieta/uso terapêutico , Método Duplo-Cego , Esquema de Medicação , Expressão Gênica , Humanos , Inflamação/tratamento farmacológico , Interleucina-10/genética , Obesidade/tratamento farmacológico , Sobrepeso/tratamento farmacológico , Proteoglicanas/administração & dosagem , Proteoglicanas/uso terapêutico , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Amido/farmacologia
17.
J Immunol ; 181(5): 3690-7, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18714045

RESUMO

Dendritic cell (DC)-based antitumor immunotherapy is a promising cancer therapy. We have previously shown that tumor-derived TGF-beta limits the efficacy of the DC/tumor fusion vaccine in mice. In the current study we investigated the effect of neutralizing tumor-derived TGF-beta on the efficacy of the DC/tumor fusion vaccine. An adenovirus encoding human TGF-beta receptor type II fused to the Fc region of human IgM (Adv-TGF-beta-R) or a control adenovirus encoding LacZ (Adv-LacZ) was used to express a soluble form of the neutralizing TGF-beta receptor (TGF-beta-R). Murine breast carcinoma cells, 4T1, but not bone marrow-derived DCs, were successfully transfected with Adv-TGF-beta-R (4T1+Adv-TGF-beta-R) using a multiplicity of infection of 300. Immunization with irradiated 4T1+Adv-TGF-beta-R tumor cells conferred enhanced antitumor immunity compared with immunization with irradiated 4T1+Adv-LacZ tumor cells. The DC/4T1+Adv-TGF-beta-R fusion vaccine offered enhanced protective and therapeutic efficacy compared with the DC/4T1-Adv-LacZ fusion vaccine. Because TGF-beta is known to induce regulatory T cells (Tregs), we further showed that the DC/4T1+Adv-TGF-beta-R fusion vaccine induced fewer CD4(+)CD25(+)Foxp3(+) Tregs than the DC/4T1+Adv-LacZ fusion vaccine in vitro and in vivo. The suppressive role of splenic CD4(+)CD25(+) Tregs isolated from mice immunized with DC/4T1+Adv-LacZ was demonstrated using a CTL killing assay. Similar enhanced therapeutic efficacy was observed in murine renal cell carcinoma, RenCa, which expresses a high level of TGF-beta. We conclude that the blockade of tumor-derived TGF-beta reduces Treg induction by the DC/tumor fusion vaccine and enhances antitumor immunity. This may be an effective strategy to enhance human DC-based antitumor vaccines.


Assuntos
Antígenos de Neoplasias , Vacinas Anticâncer , Neoplasias/terapia , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Animais , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/química , Neoplasias/patologia , Receptores de Fatores de Crescimento Transformadores beta/genética , Linfócitos T Reguladores , Transfecção
18.
Infect Immun ; 76(6): 2325-32, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18378634

RESUMO

Deletion of integrin alphav beta6 has been associated with significant protection in experiments where lung injury was induced by bleomycin, lipophilic polysaccharides, and high tidal volume ventilation. This has led to the suggestion that antibody blockade of this integrin is a novel therapy for acute lung injury. We questioned whether beta6 gene deletion would also protect against Pseudomonas aeruginosa-induced acute lung injury. Wild-type and littermate beta6-null mice, as well as recombinant soluble TGF-beta receptor type II-Fc (rsTGF-betaRII-Fc) and anti-alphav beta6 treated wild-type mice, were instilled with live P. aeruginosa. Four or 8 h after bacterial instillation, the mice were euthanized, and either bronchoalveolar lavage fluid or lung homogenates were obtained. Deletion of the beta6 gene resulted in an overall increase in inflammatory cells in the lungs. Bacterial numbers from the lung homogenates of infected beta6-null mice were significantly decreased compared to the numbers in the wild-type mice (1.6 x 10(6) CFU versus 4.2 x 10(6) CFU [P < 0.01]). There were no significant differences in P. aeruginosa-mediated increases in lung endothelial permeability between wild-type and beta6-null mice. Similarly, pretreatment with the alphav beta6 antibody or with rsTGF-betaRII-Fc did not significantly affect the P. aeruginosa-induced lung injury or rate of survival compared to pretreatment with control immunoglobulin G. We conclude that deletion or inhibition of the integrin alphav beta6 did not protect animals from P. aeruginosa-induced lung injury. However, these therapies also did not increase the lung injury, suggesting that host defense was not compromised by this promising new therapy.


Assuntos
Antígenos de Neoplasias/metabolismo , Integrinas/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/microbiologia , Animais , Anticorpos Bloqueadores/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/análise , Citocinas/metabolismo , Deleção de Genes , Regulação da Expressão Gênica , Integrinas/genética , Integrinas/imunologia , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Knockout , Fagocitose/genética , Infecções por Pseudomonas/microbiologia , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Síndrome do Desconforto Respiratório/prevenção & controle , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
19.
Exp Hematol ; 35(1): 64-74, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17198875

RESUMO

Myelofibrosis is characterized by excessive deposits of extracellular matrix proteins, which occur as a marrow microenvironment reactive response to cytokines released from the clonal malignant myeloproliferation. The observation that mice exposed to high systemic levels of thrombopoietin (TPO) invariably developing myelofibrosis has allowed demonstration of the crucial role of transforming growth factor (TGF)-beta1 released by hematopoietic cells in the onset of myelofibrosis. The purpose of this study was to investigate whether TGF-beta1 inhibition could directly inhibit fibrosis development in a curative approach of this mice model. An adenovirus encoding for TGF-beta1 soluble receptor (TGF-beta-RII-Fc) was injected either shortly after transplantation (preventive) or 30 days post-transplantation (curative). Mice were transplanted with syngenic bone marrow cells transduced with a retrovirus encoding for murine TPO. All mice developed a myeloproliferative syndrome. TGF-beta-RII-Fc was detected in the blood of all treated mice, leading to a dramatic decrease in TGF-beta1 level. Histological analysis show that the two approaches (curative or preventive) were successful enough to inhibit bone marrow and spleen fibrosis development in this model. However, lethality of TPO overexpression was not decreased after treatment, indicating that in this mice model, myeloproliferation rather than fibrosis was probably responsible for the lethality induced by the disorder.


Assuntos
Terapia Genética/métodos , Mielofibrose Primária/terapia , Receptores de Fatores de Crescimento Transformadores beta/administração & dosagem , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Adenoviridae , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Modelos Animais de Doenças , Camundongos , Camundongos SCID , Mielofibrose Primária/prevenção & controle , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Esplenopatias/terapia , Análise de Sobrevida , Trombopoetina/administração & dosagem , Trombopoetina/genética , Transdução Genética , Transplante Isogênico
20.
Diabetes ; 56(2): 380-8, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17259382

RESUMO

Transforming growth factor-beta (TGF-beta) has previously been implicated in the progression of diabetic nephropathy, including the onset of fibrosis and albuminuria. Here we report for the first time the use of a high-affinity TGF-beta1 binding molecule, the soluble human TGF-beta type II receptor (sTbetaRII.Fc), in the treatment of diabetic nephropathy in 12-week streptozotocin-induced diabetic Sprague-Dawley rats. In vitro studies using immortalized rat proximal tubule cells revealed that 50 pmol/l TGF-beta1 disrupted albumin uptake (P < 0.001 vs. control), an inhibition significantly reversed by the use of the sTbetaRII.Fc (1,200 pmol/l). In vivo studies demonstrated that treatment with sTbetaRII.Fc reduced urinary albumin excretion by 36% at 4 weeks, 59% at 8 weeks (P < 0.001), and 45% at 12 weeks (P < 0.01 for diabetic vs. treated). This was correlated with an increase in megalin expression (P < 0.05 for diabetic vs. treated) and a reduction in collagen IV expression following sTbetaRII.Fc treatment (P < 0.001 for diabetic vs. treated). These changes occurred independently of changes in blood glucose levels. This study demonstrates that the sTbetaRII.Fc is a potential new agent for the treatment of fibrosis and albuminuria in diabetic nephropathy and may reduce albuminuria by reducing TGF-beta1-induced disruptions of renal proximal tubule cell uptake of albumin.


Assuntos
Albuminúria/tratamento farmacológico , Nefropatias Diabéticas/tratamento farmacológico , Túbulos Renais Proximais/efeitos dos fármacos , Receptores de Fatores de Crescimento Transformadores beta/uso terapêutico , Fator de Crescimento Transformador beta1/metabolismo , Animais , Células Cultivadas , Colágeno Tipo IV/biossíntese , Diabetes Mellitus Experimental , Nefropatias Diabéticas/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Proteínas Serina-Treonina Quinases , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo II , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA