Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 574
Filtrar
1.
Breast Cancer Res Treat ; 192(2): 447-455, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35034243

RESUMO

PURPOSE: Black women have a 40% increased risk of breast cancer-related mortality. These outcome disparities may reflect differences in tumor pathways and a lack of targetable therapies for specific subtypes that are more common in Black women. Hepatocyte growth factor (HGF) is a targetable pathway that promotes breast cancer tumorigenesis, is associated with basal-like breast cancer, and is differentially expressed by race. This study assessed whether a 38-gene HGF expression signature is associated with recurrence and survival in Black and non-Black women. METHODS: Study participants included 1957 invasive breast cancer cases from the Carolina Breast Cancer Study. The HGF signature was evaluated in association with recurrence (n = 1251, 171 recurrences), overall, and breast cancer-specific mortality (n = 706, 190/328 breast cancer/overall deaths) using Cox proportional hazard models. RESULTS: Women with HGF-positive tumors had higher recurrence rates [HR 1.88, 95% CI (1.19, 2.98)], breast cancer-specific mortality [HR 1.90, 95% CI (1.26, 2.85)], and overall mortality [HR 1.69; 95% CI (1.17, 2.43)]. Among Black women, HGF positivity was significantly associated with higher 5-year rate of recurrence [HR 1.73; 95% CI (1.01, 2.99)], but this association was not significant in non-Black women [HR 1.68; 95% CI (0.72, 3.90)]. Among Black women, HGF-positive tumors had elevated breast cancer-specific mortality [HR 1.80, 95% CI (1.05, 3.09)], which was not significant in non-Black women [HR 1.52; 95% CI (0.78, 2.99)]. CONCLUSION: This multi-gene HGF signature is a poor-prognosis feature for breast cancer and may identify patients who could benefit from HGF-targeted treatments, an unmet need for Black and triple-negative patients.


Assuntos
Neoplasias da Mama , Fator de Crescimento de Hepatócito , População Negra , Neoplasias da Mama/etnologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Feminino , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Modelos de Riscos Proporcionais , Fatores Raciais , População Branca
2.
PLoS One ; 16(12): e0261402, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34936660

RESUMO

INTRODUCTION: An underactive bladder can lead to difficulty in voiding that causes incomplete emptying of the bladder, suggesting the need for a new strategy to increase bladder contractility in such patients. This study was performed to investigate whether human mesenchymal stem cells (hMSCs) were capable of restoring bladder contractility in rats with underactive bladder due to bladder outlet obstruction (BOO) and enhancing their effects by overexpressing hepatocyte growth factor (HGF) in hMSCs. MATERIALS AND METHODS: The hMSCs were transplanted into the bladder wall of rats. Fifty female Sprague-Dawley rats at six weeks of age were divided into five groups: group 1: control; group 2: sham intervention; group 3: eight-week BOO; group 4: BOO rats transplanted with hMSCs; and group 5: BOO rats transplanted with hMSCs overexpressing HGF. Two weeks after the onset of BOO in groups 4 and 5, hMSCs were injected into the bladder wall. Cystometry evaluation was followed by Masson's trichrome staining of bladder tissues. Realtime PCR and immunohistochemical staining were performed to determine for hypoxia, apoptosis, and angiogenesis. RESULTS: Collagen deposition of bladder increased in BOO but decreased after transplantation of hMSCs. The increased inter-contraction interval and residual urine volume after BOO was reversed after hMSCs transplantation. The decreased maximal voiding pressure after BOO was restored by hMSCs treatment. The mRNA expression of bladder collagen1 and TGF-ß1 increased in BOO but decreased after hMSCs transplantation. The decrease in vWF-positive cells in the bladder following BOO was increased after hMSCs transplantation. Caspase 3 and TUNEL-positive apoptosis of bladder cells increased in BOO but decreased after transplantation of hMSCs. These effects were enhanced by overexpressing HGF in hMSCs. CONCLUSION: Transplantation of hMSCs into bladder wall increased the number of micro-vessels, decreased collagen deposition and apoptosis of detrusor muscle, and improved bladder underactivity. The effects were enhanced by overexpressing HGF in hMSCs. Our findings suggest that the restoration of underactive bladder using hMSCs may be used to rectify micturition disorders in patients following resolution of BOO. Further studies are needed before hMSCs can be used in clinical applications.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Transplante de Células-Tronco Mesenquimais , Obstrução do Colo da Bexiga Urinária/cirurgia , Bexiga Inativa/cirurgia , Bexiga Urinária/fisiopatologia , Animais , Linhagem Celular , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Humanos , Células-Tronco Mesenquimais/fisiologia , Contração Muscular/fisiologia , Neovascularização Fisiológica/fisiologia , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia , Micção/fisiologia
3.
Exp Neurol ; 341: 113688, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33713655

RESUMO

Increasing numbers of patients with spontaneous subarachnoid hemorrhage(SAH) who recover from surgery and intensive care management still live with cognitive impairment after discharge, indicating the importance of white matter injury at the acute stage of SAH. In the present study, standard endovascular perforation was employed to establish an SAH mouse model, and a microRNA (miRNA) chip was used to analyze the changes in gene expression in white matter tissue after SAH. The data indicate that 17 miRNAs were downregulated, including miR-706, miR-669a-5p, miR-669p-5p, miR-7116-5p and miR-195a-3p, while 13 miRNAs were upregulated, including miR-6907-5p, miR-5135, miR-6982-5p, miR-668-5p, miR-8119. Strikingly, miR-706 was significantly downregulated with the highest fold change. Further experiments confirmed that miR-706 could alleviate white matter injury and improve neurological behavior, at least partially by inhibiting the PKCα/MST1/NF-κB pathway and the release of inflammatory cytokines. These results might provide a deeper understanding of the pathophysiological processes in white matter after SAH, as well as potential therapeutic strategies for the translational research.


Assuntos
Fator de Crescimento de Hepatócito/antagonistas & inibidores , MicroRNAs/biossíntese , NF-kappa B/antagonistas & inibidores , Proteína Quinase C-alfa/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Hemorragia Subaracnóidea/metabolismo , Substância Branca/metabolismo , Animais , Regulação para Baixo/fisiologia , Fator de Crescimento de Hepatócito/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/biossíntese , Proteína Quinase C-alfa/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/prevenção & controle , Substância Branca/lesões , Substância Branca/patologia
4.
Biotechnol Bioeng ; 118(1): 72-81, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32880912

RESUMO

Diseases leading to terminal hepatic failure are among the most common causes of death worldwide. Transplant of the whole organ is the only effective method to cure liver failure. Unfortunately, this treatment option is not available universally due to the serious shortage of donors. Thus, alternative methods have been developed that are aimed at prolonging the life of patients, including hepatic cells transplantation and bridging therapy based on hybrid bioartificial liver devices. Parenchymal liver cells are highly differentiated and perform many complex functions, such as detoxification and protein synthesis. Unfortunately, isolated hepatocytes display a rapid decline in viability and liver-specific functions. A number of methods have been developed to maintain hepatocytes in their highly differentiated state in vitro, amongst them the most promising being 3D growth scaffolds and decellularized tissues or coculture with other cell types required for the heterotypic cell-cell interactions. Here we present a novel approach to the hepatic cells culture based on the feeder layer cells genetically modified using lentiviral vector to stably produce additional amounts of hepatocyte growth factor and show the positive influence of these coculture conditions on the preservation of the hepatic functions of the liver parenchymal cells' model-C3A cells.


Assuntos
Fator de Crescimento de Hepatócito/biossíntese , Hepatócitos/metabolismo , Fígado/metabolismo , Modelos Biológicos , Pele/metabolismo , Linhagem Celular , Técnicas de Cocultura , Fibroblastos/citologia , Fator de Crescimento de Hepatócito/genética , Hepatócitos/citologia , Humanos , Fígado/citologia , Pele/citologia , Engenharia Tecidual
5.
Pediatr Surg Int ; 36(5): 597-602, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32200404

RESUMO

BACKGROUND: The prognosis of biliary atresia (BA) remains difficult to predict. This study evaluated the roles of hepatocyte growth factor (HGF) and its receptor (C-met) towards clinical outcome and native liver survival. METHODS: Hepatic HGF and C-met expression were determined using immunohistochemistry from liver biopsies of 41 BA patients during Kasai operation, and 17 non-cholestatic patients. The HGF and C-met expression was visually scored as per its intensity and percentage of stained area. BA patients were classified as high- and low-HGF and C-met receptor status. Native liver survival was compared between the two groups at 3-year follow-up. Data are shown as median and range. MAIN RESULTS: Median age of BA patients was 2 (1-6) months. Hepatic HGF and C-met staining scores of BA patients were higher than those of non-cholestatic patients (P < 0.0001). There was a correlation between HGF and C-met staining scores (spearman r = 0.77, P < 0.0001). However, there was no association between their expression and early outcome at 6 months post-op. Mean follow-up time was 68.6 months. Survival analysis revealed that native liver survival at 1 year and 3 years were 88% and 77%, respectively. Additionally, 82.6% (19/23) of patients in the low-HGF group survived with native liver, compared with 66.7% (10/15) of those in high-HGF group (P = 0.436). For C-met expression, 78.6% (22/28) of low-score and 70% (7/10) of high score groups survived with native liver (P = 0.673). CONCLUSIONS: Strong expression of hepatic HGF and its receptor in BA patients was demonstrated. However, the expression was not associated with the early outcome and native liver survival. These results suggest that HGF involved in the liver pathology of BA but its expression cannot be used as a prognostic indicator. Small sample size of patients was a main limitation. Further studies are warranted to validate our findings.


Assuntos
Atresia Biliar/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Fígado/metabolismo , Atresia Biliar/patologia , Biomarcadores/metabolismo , Biópsia , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Lactente , Transplante de Fígado , Masculino , Portoenterostomia Hepática , Prognóstico , Fatores de Tempo
6.
Oxid Med Cell Longev ; 2019: 4826525, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781338

RESUMO

Peroxisome proliferator-activated receptor- (PPAR-) γ is a ligand-dependent transcription factor, and it has become evident that PPAR-γ agonists have renoprotective effects, but their influence and mechanism during the development of calcium oxalate (CaOx) nephrolithiasis remain unknown. Rosiglitazone (RSG) was used as a representative PPAR-γ agonist in our experiments. The expression of transforming growth factor-ß1 (TGF-ß1), hepatocyte growth factor (HGF), c-Met, p-Met, PPAR-γ, p-PPAR-γ (Ser112), Smad2, Smad3, pSmad2/3, and Smad7 was examined in oxalate-treated Madin-Darby canine kidney (MDCK) cells and a stone-forming rat model. A CCK-8 assay was used to evaluate the effects of RSG on cell viability. In addition, intracellular reactive oxygen species (ROS) levels were monitored, and lipid peroxidation in renal tissue was detected according to superoxide dismutase and malondialdehyde levels. Moreover, the location and extent of CaOx crystal deposition were evaluated by Pizzolato staining. Our results showed that, both in vitro and in vivo, oxalate impaired PPAR-γ expression and phosphorylation, and then accumulative ROS production was observed, accompanied by enhanced TGF-ß1 and reduced HGF. These phenomena could be reversed by the addition of RSG. RSG also promoted cell viability and proliferation and decreased oxidative stress damage and CaOx crystal deposition. However, these protective effects of RSG were abrogated by the PPAR-γ-specific inhibitor GW9662. Our results revealed that the reduction of PPAR-γ activity played a critical role in oxalate-induced ROS damage and CaOx stone formation. RSG can regulate TGF-ß1 and HGF/c-Met through PPAR-γ to exert antioxidant effects against hyperoxaluria and alleviate crystal deposition. Therefore, PPAR-γ agonists may be expected to be a novel therapy for nephrolithiasis, and this effect is related to PPAR-γ-dependent suppression of oxidative stress.


Assuntos
Oxalato de Cálcio/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/biossíntese , Rim/metabolismo , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Rosiglitazona/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Animais , Cães , Células Epiteliais/patologia , Hiperoxalúria/tratamento farmacológico , Hiperoxalúria/metabolismo , Hiperoxalúria/patologia , Rim/patologia , Células Madin Darby de Rim Canino , Masculino , Nefrolitíase/tratamento farmacológico , Nefrolitíase/metabolismo , Nefrolitíase/patologia , Ratos Sprague-Dawley
7.
Cancer Control ; 26(1): 1073274819875736, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31533461

RESUMO

Cyclin E and hepatocyte growth factor (HGF) have been observed as a multifaceted factor in many cancers, and the assessment of microvascular density (MVD) and micro-lymphatic vessel density (MLVD) has been used to quantify tumor angiogenesis and lymphangiogenesis. The aim of this study was to explore the association between expression of cyclin E, HGF, MVD, and MLVD, and clinicopathologic parameters in esophageal squamous cell carcinoma (ESCC). The expression of cyclin E, HGF, MVD, and MLVD were detected using immunohistochemically anticyclin E, HGF, CD34, and lymphatic vessel endothelial hyaluronan receptor 1 in 168 surgically resected ESCC cases and 30 normal esophageal mucosal samples. The expression levels of cyclin E, HGF, MVD, and MLVD were higher compared to controls. High cyclin E and HGF expression was found more frequently in the tumors larger than 5 cm (P < .001), with poorer differentiation (P = .034) and higher tumor node metastasis (TNM) staging (P = .009) compared to their counterparts. Both MVD and MLVD values were found to be higher in the tumors larger than 5 cm (P < .001), with poorer differentiation (P < .001) and higher TNM staging (P < .001) compared to their counterparts. Furthermore, the expression of MVD and MLVD in both the high cyclin E and high HGF expression groups was significantly higher compared to the low cyclin E and HGF expression groups (P < .001). This study demonstrated that high cyclin E and HGF expression is closely correlated with tumor size, tumor differentiation degree, and TNM stage in patients with ESCC. These findings proposed that cyclin E and HGF could serve as novel molecular markers for preoperational evaluation of ESCC.


Assuntos
Biomarcadores Tumorais/biossíntese , Carcinoma de Células Escamosas/metabolismo , Ciclina E/biossíntese , Neoplasias Esofágicas/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Vasos Linfáticos/metabolismo , Neovascularização Patológica/metabolismo , Idoso , Carcinoma de Células Escamosas/irrigação sanguínea , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/irrigação sanguínea , Neoplasias Esofágicas/patologia , Feminino , Humanos , Imuno-Histoquímica , Vasos Linfáticos/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Prognóstico
8.
Cell Transplant ; 28(9-10): 1132-1139, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31304790

RESUMO

Multilineage-differentiating stress-enduring (Muse) cells are endogenous pluripotent stem cells that can be isolated based on stage-specific embryonic antigen-3 (SSEA-3), a pluripotent stem cell-surface marker. However, their capacities for survival, neurotrophic factor secretion, and neuronal and glial differentiation are unclear in rodents. Here we analyzed mouse adipose tissue-derived Muse cells in vitro. We collected mesenchymal stem cells (MSCs) from C57BL/6 J mouse adipose tissue and separated SSEA-3+, namely Muse cells, and SSEA-3-, non-Muse cells, to assess self-renewability; pluripotency marker expression (Nanog, Oct3/4, Sox2, and SSEA-3); spontaneous differentiation into endodermal, mesodermal, and ectodermal lineages; and neural differentiation capabilities under cytokine induction. Neurally differentiated Muse and non-Muse cell functions were assessed by calcium imaging. Antioxidant ability was measured to assess survival under oxidative stress. Brain-derived neurotrophic factor (BDNF), vascular endothelial cell growth factor (VEGF), and hepatocyte growth factor (HGF) secretion were analyzed in enzyme-linked immunosorbent assays. SSEA-3+ Muse cells (6.3 ± 1.9% of mouse adipose-MSCs), but not non-Muse cells, exhibited self-renewability, spontaneous differentiation into the three germ layers, and differentiation into cells positive for Tuj-1 (27 ± 0.9%), O4 (17 ± 3.4%), or GFAP (23 ± 1.3%) under cytokine induction. Neurally differentiated Muse cells responded to KCl depolarization with greater increases in cytoplasmic Ca2+ levels than non-Muse cells. Cell survival under oxidative stress was significantly higher in Muse cells (50 ± 2.7%) versus non-Muse cells (22 ± 2.8%). Muse cells secreted significantly more BDNF, VEGF, and HGF (273 ± 12, 1479 ± 7.5, and 6591 ± 1216 pg/mL, respectively) than non-Muse cells (133 ± 4.0, 1165 ± 20, and 2383 ± 540 pg/mL, respectively). Mouse Muse cells were isolated and characterized for the first time. Muse cells showed greater pluripotency-like characteristics, survival, neurotrophic factor secretion, and neuronal and glial-differentiation capacities than non-Muse cells, indicating that they may have better neural-regeneration potential.


Assuntos
Tecido Adiposo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Diferenciação Celular , Fator de Crescimento de Hepatócito/biossíntese , Células-Tronco Mesenquimais/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Tecido Adiposo/citologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Feminino , Células-Tronco Mesenquimais/citologia , Camundongos , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Estresse Oxidativo
9.
Int J Mol Sci ; 20(12)2019 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-31238604

RESUMO

Cell therapy remains a promising approach for the treatment of cardiovascular diseases. In this regard, the contemporary trend is the development of methods to overcome low cell viability and enhance their regenerative potential. In the present study, we evaluated the therapeutic potential of gene-modified adipose-derived stromal cells (ADSC) that overexpress hepatocyte growth factor (HGF) in a mice hind limb ischemia model. Angiogenic and neuroprotective effects were assessed following ADSC transplantation in suspension or in the form of cell sheet. We found superior blood flow restoration, tissue vascularization and innervation, and fibrosis reduction after transplantation of HGF-producing ADSC sheet compared to other groups. We suggest that the observed effects are determined by pleiotropic effects of HGF, along with the multifactorial paracrine action of ADSC which remain viable and functionally active within the engineered cell construct. Thus, we demonstrated the high therapeutic potential of the utilized approach for skeletal muscle recovery after ischemic damage associated with complex tissue degenerative effects.


Assuntos
Tecido Adiposo/citologia , Fator de Crescimento de Hepatócito/biossíntese , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/metabolismo , Células Estromais/metabolismo , Células Estromais/transplante , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Movimento Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Humanos , Isquemia , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Crescimento Neuronal/efeitos dos fármacos
10.
Cell Physiol Biochem ; 52(4): 935-950, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30964610

RESUMO

BACKGROUND/AIMS: Although mesenchymal stem cells (MSCs) provide effective therapy for liver fibrosis, there are conflicting data regarding their marginal therapeutic effects. This study aimed to enhance the potential of hepatocyte regeneration in human adipose mesenchymal stem cells (ASCs) and investigate whether they have robust therapeutic efficacy in experimental liver fibrosis. METHODS: ASCs were cultured with four cytokines (ASC-C), the expression of hepatogenic factors was detected by microarray, and the effects of conditioned medium (CM) from ASC-C on the activation of hepatic stellate cells were analyzed. The therapeutic effects and mechanism of liver fibrosis induced by thioacetamide (TAA) were determined after cell transplantation. RESULTS: ASC-C exhibited high levels of hepatogenic (HGF, G-CSF), anti-apoptotic (IGFBP-2), and chemokine (IL-8) genes and increased expression of hepatocyte specific proteins. ASC-C CM inhibited the activation of hepatic stellate cells in vitro, and injection of ASC-C significantly delayed TAA-induced liver fibrosis and improved liver function and regeneration in vivo. In addition, human albumin-expressing ASC-C were observed in the livers of recipient animals. High levels of expression of HGF and its downstream signaling molecules, including p-38, were detected in the ASC-C-injected livers. Transplantation of ASC-C exerts anti-fibrotic effects and accelerates liver regeneration. CONCLUSION: Thus, ASC-C may be a novel candidate for the enhanced treatment of liver cirrhosis in clinical settings.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Adipócitos/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Sistema de Sinalização das MAP Quinases , Transplante de Células-Tronco , Células-Tronco/metabolismo , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/terapia , Adipócitos/patologia , Animais , Linhagem Celular , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco/patologia
11.
J Biochem Mol Toxicol ; 33(6): e22316, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30897285

RESUMO

The system of hepatocyte growth factor (HGF) and its receptor c-Met plays a critical role in tumor invasive growth and metastasis. The mortality rate of colorectal cancer (CRC), one of the most commonly diagnosed malignancies, is increased by it gradual development into metastasis, most frequently in the liver. Overexpression of c-Met, the protein tyrosine kinase receptor for the HCF/scatter factor, has been implicated in the progression and metastasis of human colorectal carcinoma. In this study, we aimed to investigate the role of c-Met in CRC liver metastasis and illustrate the clinical impact of regulating HGF/c-Met signaling in patients with CRC liver metastasis. We found that (I) higher levels of c-Met expression (mRNA and Protein) in CRC liver metastasis than primary CRC by assessing the patient tissue samples; (II) a positive correlation of c-Met expression with tumor stages of CRC liver metastasis, as well as c-Met expression in CRC, live metastasis concurred with regional lymph node metastasis; (III) the clinical impact of downregulation of HGF/c-Met signaling on the reduction of proliferation and invasion in CRC liver metastasis. Therefore, we demonstrate that the regulation of HGF/c-Met pathways may be a promising strategy in the treatment of patients with CRC liver metastasis.


Assuntos
Neoplasias Colorretais , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/biossíntese , Neoplasias Hepáticas , Fígado/metabolismo , Proteínas Proto-Oncogênicas c-met/biossíntese , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Feminino , Humanos , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/secundário , Neoplasias Hepáticas/terapia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica
12.
Tissue Eng Part A ; 25(1-2): 121-130, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717619

RESUMO

RNA-based vector delivery is a promising gene therapy approach. Recent advances in chemical modification of mRNA structure to form modified mRNA (mmRNA or cmRNA or modRNA) have substantially improved their stability and translational efficiency within cells. However, mmRNA conventionally delivered in solution can be taken up nonspecifically or become cleared away prematurely, which markedly limits the potential benefit of mmRNA therapy. To address this limitation, we developed mmRNA-incorporated nanofibrillar scaffolds that could target spatially localized delivery and temporally controlled release of the mmRNA both in vitro and in vivo. To establish the efficacy of mmRNA therapy, mmRNA encoding reporter proteins such as green fluorescence protein or firefly luciferase (Fluc) was loaded into aligned nanofibrillar collagen scaffolds. The mmRNA was released from mmRNA-loaded scaffolds in a transient and temporally controlled manner and induced transfection of human fibroblasts in a dose-dependent manner. In vitro transfection was further verified using mmRNA encoding the angiogenic growth factor, hepatocyte growth factor (HGF). Finally, scaffold-based delivery of HGF mmRNA to the site of surgically induced muscle injury in mice resulted in significantly higher vascular regeneration after 14 days, compared to implantation of Fluc mmRNA-releasing scaffolds. After transfection with Fluc mmRNA-releasing scaffold in vivo, Fluc activity was detectable and localized to the muscle region, based on noninvasive bioluminescence imaging. Scaffold-based local mmRNA delivery as an off-the-shelf form of gene therapy has broad translatability for treating a wide range of diseases or injuries.


Assuntos
Colágeno , Fator de Crescimento de Hepatócito , Nanofibras/química , RNA Mensageiro , Transfecção/métodos , Linhagem Celular , Colágeno/química , Colágeno/farmacologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacologia , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Humanos , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia
13.
J Cell Biochem ; 120(3): 3637-3650, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30242894

RESUMO

Mesenchymal stem cells (MSCs) protect the endothelial barrier complex and survival, implicated in the pathogenesis of acute lung injury (ALI) via paracrine hepatocyte growth factor (HGF). However, the mechanism of HGF in endothelial regulation remains unclear. Here, we introduced a coculture protocol of pulmonary microvascular endothelial cells (PMVECs) and overexpression of the HGF gene of MSCs (MSC-HGF). Immunofluorescence and endothelial permeability analysis revealed that MSC-HGF protected endothelial tight junction protein occludin expression and attenuated cellular permeability as well as endothelial apoptosis. To investigate the novel mechanism mammalian TOR (mTOR)/ signal transducer and activator of transcription 3 (STAT-3) signaling in HGF protective effects against endothelial barrier and apoptosis, we used recombinant mouse HGF in endothelial cells. In addition, we used mTOR inhibitor rapamycin to inhibit the mTOR pathway. Our study demonstrated that rapamycin decreased the protective effects of HGF on the endothelium by decreasing tight junction protein occludin expression and cell proliferation, and raising lipopolysaccharide (LPS)-induced endothelial permeability, endothelial cell injury factors ET-1 and vWF. Similarly, the protective effects of HGF on reducing endothelial barrier and apoptosis were weakened when PMVECs were treated with the STAT-3 inhibitor S3I-201. Moreover, mTOR/STAT-3 were activated by HGF demonstrated as raising mTOR (Ser2448) and STAT3 (Ser727) phosphorylation proteins, leading to endothelial barrier improvement and survival. Reversely, rapamycin or S3I-201 inhibited mTOR/STAT-3 activation. Taken together, our findings highlight that the activation of the mTOR/STAT-3 pathway provides novel mechanistic insights into MSC-secreted HGF protection against LPS-induced vascular endothelial permeability dysfunction and apoptosis, which contributes to decreasing microvascular loss and lung injury.


Assuntos
Apoptose/efeitos dos fármacos , Permeabilidade Capilar/efeitos dos fármacos , Células Endoteliais/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Lipopolissacarídeos/toxicidade , Células-Tronco Mesenquimais/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Células Endoteliais/patologia , Células-Tronco Mesenquimais/patologia , Camundongos
14.
J Interferon Cytokine Res ; 39(3): 147-154, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30592626

RESUMO

Liver sinusoidal endothelial cells (LSECs) are highly specialized and involved in hepatic regeneration by interacting with hepatic stellate cells (HSCs) and hepatocytes in a paracrine manner. However, hepatic injury can impair cellular activity and lead to endothelial dysfunction, eventually inducing the development of critical hepatic disease, including cirrhosis. Because LSECs exert their effects through paracrine factors, maintenance of paracrine potentials and survival activity in LSECs under injury stress is a critical strategy for inhibiting disease progression. This study explored the effect of Substance-P (SP) on cell viability, proliferation, and nitric oxide (NO)/hepatocyte growth factor (HGF) production in LSECs. Under noninjured conditions, SP treatment enhanced cell viability, cell proliferation, and HGF/NO secretion in LSECs. In the presence of tumor necrosis factor (TNF)-α-induced inflammatory stress, SP blocked TNF-α-induced endothelial dysfunction, accompanied by elevated cell viability and NO/HGF secretion. Interestingly, SP-primed LSEC-conditioned medium accelerated hepatocyte repopulation without causing morphological alterations. The primitive effect of SP was reversed by endothelial nitric oxide synthase inhibitor or HGF/c-MET inhibitor, indicating the importance of the NO/HGF combination in hepatic regeneration by SP. Taken together, these results suggest SP can protect LSECs from inflammatory stress by NO/HGF, which contributes to hepatocyte repopulation.


Assuntos
Células Endoteliais/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Fígado/metabolismo , Óxido Nítrico/biossíntese , Substância P/metabolismo , Sobrevivência Celular , Células Hep G2 , Fator de Crescimento de Hepatócito/análise , Humanos , Fígado/patologia , Óxido Nítrico/análise
15.
Physiol Rep ; 6(16): e13794, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30155985

RESUMO

TGF beta is a multifunctional cytokine that is important in the pathogenesis of pulmonary fibrosis. The ability of TGF beta to stimulate smooth muscle actin and extracellular matrix gene expression in fibroblasts is well established. In this report, we evaluated the effect of TGF beta on the expression of HGF, FGF7 (KGF), and FGF10, important growth and survival factors for the alveolar epithelium. These growth factors are important for maintaining type II cells and for restoration of the epithelium after lung injury. Under conditions of normal serum supplementation or serum withdrawal TGF beta inhibited fibroblast expression of HGF, FGF7, and FGF10. We confirmed these observations with genome wide RNA sequencing of the response of control and IPF fibroblasts to TGF beta. In general, gene expression in IPF fibroblasts was similar to control fibroblasts. Reduced expression of HGF, FGF7, and FGF10 is another means whereby TGF beta impairs epithelial healing and promotes fibrosis after lung injury.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibrose Pulmonar Idiopática/patologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Fator de Crescimento Transformador beta/farmacologia , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Meios de Cultura Livres de Soro , Feminino , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fator 7 de Crescimento de Fibroblastos/biossíntese , Fator 7 de Crescimento de Fibroblastos/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/genética , Fator de Crescimento Transformador beta/fisiologia
16.
Toxicol Appl Pharmacol ; 343: 1-15, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29408570

RESUMO

Berberine has been demonstrated to alleviate renal interstitial, liver and myocardial fibrosis when administered orally despite its extremely low bioavailability. Here, we inspected effect of berberine on pulmonary fibrosis (PF) and explored underlying mechanisms on the basis of intestinal endocrine. The results showed that either oral or rectal administration of berberine exhibited marked alleviation of bleomycin-induced PF in mice. In contrast, anti-PF activity of berberine disappeared when given by an intravenous injection, implying that it functioned in a gut-dependent manner. Moreover, berberine promoted both mRNA and protein levels of HGF and PTEN in colons, but only their protein levels in lungs of PF mice. In addition, SU11274 but not BPV abolished the anti-PF effect of berberine. In vitro, berberine preferentially induced expression of HGF in fibroblast cells than epithelial, preadipocyte and endothelial cells. Similarly, rosiglitazone and 15dPGJ2 also enhanced expression of HGF in fibroblasts cells, and GW9662 and siPPAR-γ diminished induction of berberine on HGF expression. Berberine could enter into the cytoplasm, activate PPAR-γ directly and synergistically with 15dPGJ2, as shown by an up-regulation of CD36 and aP2 mRNA expression, nuclear translocation and DNA-binding activity of PPAR-γ both in vitro and in vivo. Additionally, GW9662 almost abolished anti-PF effect of berberine and induction of HGF expression in colons. In conclusion, oral administration of berberine displays anti-PF action probably in a colon-dependent manner, and mechanisms involve activation of PPAR-γ and resultant promotion of HGF expression in colonic fibroblasts. The up-regulated HGF arrives in lung tissues via blood circulation to palliate PF.


Assuntos
Berberina/administração & dosagem , Bleomicina/toxicidade , Colo/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , PPAR gama/metabolismo , Fibrose Pulmonar/metabolismo , Células 3T3 , Administração Oral , Animais , Antibióticos Antineoplásicos/toxicidade , Colo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos ICR , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico
17.
J Am Soc Nephrol ; 29(4): 1257-1271, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29343518

RESUMO

AKI is a devastating condition with high morbidity and mortality. The pathologic features of AKI are characterized by tubular injury, inflammation, and vascular impairment. Whether fibroblasts in the renal interstitium have a role in the pathogenesis of AKI is unknown. In this study, we investigated the role of fibroblast-specific ß-catenin signaling in dictating the outcome of AKI, using conditional knockout mice in which ß-catenin was specifically ablated in fibroblasts (Gli1-ß-cat-/-). After ischemia-reperfusion injury (IRI), Gli1-ß-cat-/- mice had lower serum creatinine levels and less morphologic injury than Gli1-ß-cat+/+ littermate controls. Moreover, we detected fewer apoptotic cells, as well as decreased cytochrome C release; reduced expression of Bax, FasL, and p53; and increased phosphorylation of Akt, in the Gli1-ß-cat-/- kidneys. Gli1-ß-cat-/- kidneys also exhibited upregulated expression of proliferating cell nuclear antigen and Ki-67, which are markers of cell proliferation. Furthermore, Gli1-ß-cat-/- kidneys displayed suppressed NF-κB signaling and cytokine expression and reduced infiltration of inflammatory cells. Notably, loss of ß-catenin in fibroblasts induced renal expression of hepatocyte growth factor (HGF) and augmented the tyrosine phosphorylation of c-met receptor after IRI. In vitro, treatment with Wnt ligands or ectopic expression of active ß-catenin inhibited HGF mRNA and protein expression and repressed HGF promoter activity. Collectively, these results suggest that fibroblast-specific ß-catenin signaling can control tubular injury and repair in AKI by modulating HGF expression. Our studies uncover a previously unrecognized role for interstitial fibroblasts in the pathogenesis of AKI.


Assuntos
Injúria Renal Aguda/fisiopatologia , Fibroblastos/metabolismo , Rim/irrigação sanguínea , Traumatismo por Reperfusão/fisiopatologia , Via de Sinalização Wnt , beta Catenina/fisiologia , Injúria Renal Aguda/genética , Animais , Apoptose , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Divisão Celular , Movimento Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Fibroblastos/patologia , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/fisiologia , Inflamação , Túbulos Renais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Pirimidinonas/farmacologia , Regeneração , Traumatismo por Reperfusão/genética , beta Catenina/antagonistas & inibidores , beta Catenina/deficiência , beta Catenina/genética
18.
Circ J ; 82(5): 1459-1465, 2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28931787

RESUMO

BACKGROUND: We previously identified circulating mesoangioblasts (cMABs), a subset of mesenchymal stem cells that express cardiac mesodermal markers, in patients undergoing cardiac surgery with cardiopulmonary bypass (CPB). We also found that hepatocyte growth factor (HGF) is upregulated during cardiac surgery with CPB in humans, and induces MAB-like cell mobilization in rodents. These results strongly suggest that heparin induced MAB mobilization via HGF upregulation. Here, we tested this hypothesis in patients undergoing cardiac surgery or cardiac catheterization. We also examined whether human cMABs are derived from the heart.Methods and Results:Plasma HGF levels were determined by ELISA. Mononuclear cells isolated from blood samples were cultured on fibronectin-coated dishes, and outgrowing cMAB colonies were counted. We first confirmed that HGF upregulation and cMAB mobilization were observed before the start of CPB, excluding the possibility that CPB is the primary inducer of cMAB mobilization. We then examined patients undergoing cardiac catheterization and found that heparin significantly increased plasma HGF levels and the number of cMAB colonies in a dose-dependent manner. The results of simultaneous blood sampling from the aortic sinus, coronary sinus, and right atrium were consistent with the notion that human cMABs are derived from the heart. CONCLUSIONS: Human cMABs are mobilized by heparin injection during cardiac surgery or cardiac catheterization, presumably via HGF upregulation.


Assuntos
Cateterismo Cardíaco , Ponte Cardiopulmonar , Heparina/administração & dosagem , Fator de Crescimento de Hepatócito/biossíntese , Células-Tronco Mesenquimais/metabolismo , Idoso , Idoso de 80 Anos ou mais , Feminino , Átrios do Coração/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade
19.
Respir Res ; 18(1): 118, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28619066

RESUMO

BACKGROUND: The influence of lung fibroblasts on lung cancer progression is not fully understood. METHODS: Lung fibroblasts (HFL1, MRC5, and IMR90 cells) and non-small cell lung cancer (NSCLC)-derived cell lines (A549, EBC1, and HI1017) were cultured under serum-free conditions, and the resulting culture media were designated "cell-conditioned media". Cell survival (viability) was assessed by WST-1 assay. Concentrations of hepatocyte growth factor (HGF) were measured by ELISA. The BALB/c-nu mouse strain was used for the xenograft model. RESULTS: Lung fibroblast-conditioned media enhanced the survival of the three NSCLC cell lines tested. HGF was produced to a greater extent by lung fibroblasts than NSCLC cells. Exogenous HGF enhanced the survival of NSCLC cells. Either an anti-HGF neutralizing antibody or the Met inhibitor PHA-665752 inhibited the fibroblast-conditioned media-enhanced survival of NSCLC cells. The co-inoculation of mice with NSCLC cells and fibroblasts enhanced tumorigenicity and tumor progression in a mouse xenograft model. PHA-665752 significantly inhibited tumor progression that occurred after the co-inoculation of NSCLC cells and fibroblasts. In addition, HGF production by fibroblasts was stimulated by NSCLC cells. CONCLUSIONS: The current study provides evidence for an interaction between fibroblasts and NSCLC cells via the HGF/Met signaling pathway, which affects NSCLC cell survival and tumor progression. These findings may contribute to the development of anti-cancer-associated fibroblast therapeutic strategies. TRIAL REGISTRATION: No trial registration is required because this study is not a clinical trial. This study does not include any participants or patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fibroblastos/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Neoplasias Pulmonares/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Fibroblastos/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Mol Cell Biol ; 37(18)2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28652266

RESUMO

Long noncoding RNAs play a pivotal role in tumor progression, but their role in cancer cells in the nutrient-starved tumor microenvironment remains unknown. Here, we show that a nutrient starvation-responsive long noncoding RNA, JHDM1D antisense 1 (JHDM1D-AS1), promotes tumorigenesis by regulating angiogenesis in response to nutrient starvation. Expression of JHDM1D-AS1 was increased in cancer cells. In addition, expression of JHDM1D-AS1 was increased in clinical tumor samples compared to that in normal tissue. Stable expression of JHDM1D-AS1 in human pancreatic cancer (PANC-1 and AsPC-1) cells promoted cell growth in vitro Remarkably, these JHDM1D-AS1-expressing cells showed a significant increase in tumor growth in vivo that was associated with increased formation of CD31+ blood vessels and elevated infiltration of CD11b+ macrophage lineage cells into tumor tissues. Genome-wide analysis of tumor xenografts revealed that expression of genes for tumor-derived angiogenic factors such as hHGF and hFGF1 concomitant with host-derived inflammation-responsive genes such as mMmp3, mMmp9, mS100a8, and mS100a9 was increased in tumor xenografts of JHDM1D-AS1-expressing pancreatic cancer cells, leading to a poor prognosis. Our results provide evidence that increased JHDM1D-AS1 expression under nutrient starvation accelerates tumor growth by upregulating angiogenesis, thus laying the foundation for improved therapeutic strategies.


Assuntos
Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias/patologia , Neovascularização Patológica/genética , Inanição/genética , Animais , Calgranulina A/biossíntese , Calgranulina B/biossíntese , Linhagem Celular Tumoral , Proliferação de Células , Fator 1 de Crescimento de Fibroblastos/biossíntese , Perfilação da Expressão Gênica , Fator de Crescimento de Hepatócito/biossíntese , Humanos , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias/genética , Interferência de RNA , RNA Longo não Codificante/biossíntese , RNA Longo não Codificante/genética , RNA Interferente Pequeno/genética , Transplante Heterólogo , Microambiente Tumoral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA