Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Cancer Med ; 12(13): 14149-14156, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37325945

RESUMO

OBJECTIVE: To investigate the efficacy of tyrosine kinase inhibitors (TKIs) in the treatment of metastatic renal cell carcinoma (mRCC) with rhabdoid (mRCC-R) and sarcomatoid (mRCC-S) differentiations. MATERIALS AND METHODS: In this single-institutional cohort study, we included patients with RCC with rhabdoid (RCC-R) and sarcomatoid (RCC-S) differentiation, who were treated with TKIs after metastasis at our institute from 2013 to 2021. Patient characteristics, treatments, and clinical outcomes were recorded and analyzed. RESULTS: We identified 111 patients with RCC-R or RCC-S differentiations, of which 23 patients were included in the final analysis. Of the 23 patients, 10 (43.5%) were grouped as mRCC-R and 13 (56.5%) as mRCC-S. At a median follow-up of 40 months, mRCC-R and mRCC-S progressed in 7 of 10 and 12 of 13 patients, respectively. In addition, four and eight patients died in the mRCC-R and mRCC-S groups, respectively. The median progression-free survival (PFS) of the two groups was 19 months (mRCC-R: 95% confidence interval [CI] 4.08-33.92) and 7 months (mRCC-S: 95% CI 2.03-11.96), while the median overall survival (OS) was 32 months and 21 months, respectively. mRCC-S had a worse prognosis than mRCC-R. Based on the univariate Cox regression model, single metastasis or multiple metastasis of tumor, rhabdoid differentiation, and sarcomatoid differentiation were predictors of PFS but not OS. CONCLUSION: The efficacy of TKIs in the treatment of mRCC-R and mRCC-S may be different.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Estudos de Coortes , Prognóstico , Estudos Retrospectivos , Inibidores de Proteínas Quinases/uso terapêutico
2.
Cancer Sci ; 113(2): 733-743, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34859546

RESUMO

Breast cancer is the leading cause of cancer death among women and almost all of the breast cancer-caused mortality is related to metastasis. It has been reported that glucocorticoid facilitates the metastasis of breast cancer in mice, and mifepristone can antagonize the effect of glucocorticoid. Paclitaxel is one of the important drugs in the treatment of breast cancer. Mifepristone combined with paclitaxel could be an effective strategy for inhibiting breast cancer metastasis. However, their inherent defects, in terms of short blood circulation half-life and lack of tumor targeting, not only limit their effectiveness but also cause adverse reactions. Therefore, our aim is to explore a novel protocol against breast cancer metastasis, further optimize its therapeutic efficacy by a nanodelivery system, and explore its mechanism. Herein, a paclitaxel-conjugated and mifepristone-loaded hydrogel (PM-nano) was prepared by self-assembly. Its characterizations were studied. The antimetastatic effect was evaluated in vitro and in vivo and its mechanism was also explored by western blot assay. The resultant PM-nano was developed with favorable water solubility and good biocompatibility. Moreover, PM-nano displayed increased cell uptake properties and stimulated drug release in the tumor micro-acidic environment. The PM-nano was more effective in inhibiting the proliferation and metastasis of breast cancer than other groups in vitro and in vivo. The PM-nano might inhibit metastasis through glucocorticoid receptor/receptor tyrosine kinase-like orphan receptor 1 and MMPs. Taken together, PM-nano showed superior antimetastatic effects against breast cancer and excellent biocompatibility in vitro and in vivo, providing a new option for limiting metastasis.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Hidrogéis/uso terapêutico , Mifepristona/uso terapêutico , Paclitaxel/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Disponibilidade Biológica , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Portadores de Fármacos/uso terapêutico , Liberação Controlada de Fármacos , Feminino , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Mifepristona/química , Mifepristona/farmacologia , Nanoestruturas/uso terapêutico , Paclitaxel/química , Paclitaxel/farmacologia , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Biomed Res Int ; 2021: 8249293, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34805404

RESUMO

BACKGROUND: Pancreatic cancer is a malignant tumor of the digestive tract, which is difficult to diagnose and treat due to bad early diagnosis. We aimed to explore the role of kinesin superfamily 4A (KIF4A) in pancreatic ductal adenocarcinoma (PDAC). METHODS: We first used the bioinformatic website to screen the data of pancreatic cancer in TCGA, and KIF4A protein was detected among the 86 specimens of patients in our hospital combined with clinic-pathological characteristics and survival analysis. KIF4A loss-expression cell lines were established by RNA interference (RNAi). In addition, we performed in vitro cell assays to detect the changes in cell proliferation, migration, and invasion. The proteins involved in the proliferation and metastasis of cancer cells were also detected by western blot. The above results could be proved in vivo. Further, the correlation between KIF4A and CDC5L was analyzed by TCGA and IHC data. RESULTS: We first found a high expression of KIF4A in pancreatic cancer, suggesting a role of KIF4A in the development of pancreatic cancer. KIF4A was found to be differentially expressed (P < 0.05) among the 86 specimens of patients in our hospital and was significantly associated with PDAC TNM stages and tumor size. High KIF4A expression also significantly worsened overall survival (OS) and disease-free survival rate (DFS) (P < 0.05, respectively). In addition, cell proliferation, migration, and invasion were inhibited by the KIF4A-shRNA group compared with the control (P < 0.05, respectively). In the end, knockdown of KIF4A could inhibit tumor development and metastasis in vivo. Further, the positive correlation between KIF4A and CDC5L existed, and KIF4A might promote pancreatic cancer proliferation by affecting CDC5L expression. CONCLUSION: In conclusion, the high expression level of KIF4A in PDAC was closely related to poor clinical and pathological status, lymphatic metastasis, and vascular invasion. KIF4A might be involved in promoting the development of PDAC in vitro and in vivo, which might be a new therapeutic target of PDAC.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Cinesinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Biologia Computacional , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Estudos Retrospectivos , Regulação para Cima
4.
J Oncol ; 2021: 5572402, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539784

RESUMO

KIFs have been reported to play a critical role in a variety of tumors, and KIF20B is a protein in KFIs. In this research, KIF20B was highly expressed in the GEO database and our hospital's data, and high expression of KIF20B suggested poor prognosis. We detect the expression of KIF20B in pancreatic cancer and adjacent normal tissues using immunohistochemistry. Knockdown of KIF20B in pancreatic cancer cell lines, PANC-1 and BxPC-3 cells, inhibited cell proliferation which are detected by colony formation assays, CCK8, and western bolt of Ki-67 and PCNA. Xenograft assay showed a similar result in vivo. KIF20B is a potential therapeutic target in pancreatic cancer.

5.
Acta Biomater ; 128: 209-221, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33878473

RESUMO

The therapeutic effectiveness of cell transplantation in treatment of diseases and injuries is often limited by low cell retention, survivability, and engraftment. Extracellular matrix (ECM)-derived scaffolds are capable of controlling cell responses, thereby offering potential solutions to current challenges associated with cell therapy. However, it remains a technical challenge to produce ECM scaffolds with highly interconnected porous structure specifically required for cell transplantation. Here, we developed inverse opal porous extracellular matrix (ioECM) scaffolds through subcutaneous implantation of sacrificial templates assembled from polymer microspheres, followed by removal of the microsphere template and cellular content. Such highly interconnected porous ioECM scaffolds supported the anchorage, survival, viability, anti-apoptotic and paracrine activities of rat bone marrow mesenchymal stem cells (BMSCs), which further promoted endothelial cell migration and tube formation and viability. Upon transplantation into nude mouse critical limb ischemic model, ioECM promoted the engraftment of laden BMSCs, facilitated interconnected vascular network formation with accelerated recovery of blood perfusion and inhibited muscle atrophy and fibrosis. Our study demonstrates a unique strategy to engineer highly porous yet well-interconnected ECM scaffolds specifically for cell transplantation with marked improvement of survivability and vascularization, which offers an essential step toward the success of cell therapy and regenerative medicine. STATEMENT OF SIGNIFICANCE: Cell-based therapy has a good developing foreground applied in a variety of tissue regeneration. Extracellular matrix (ECM) scaffolds is an optimal choice for cell delivery duo to its superior biocompatibility and favorable immune responses. However, the current ECM scaffolds lacking of the controllable pore structure restrict the cell delivery efficiency and therapeutic outcome. Here, we fabricated highly interconnected inverse opal extracellular matrix (ioECM) scaffolds, which can enhance the effect of stem cell therapy in limb ischemic model by improving the survival, viability, and paracrine activities of stem cells. Our study provides reference value for the design and fabrication of ECM based biomaterials for cell transplantation.


Assuntos
Células-Tronco Mesenquimais , Alicerces Teciduais , Animais , Matriz Extracelular , Isquemia/terapia , Camundongos , Ratos , Transplante de Células-Tronco , Engenharia Tecidual
6.
J Am Soc Nephrol ; 31(10): 2292-2311, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32769144

RESUMO

BACKGROUND: Progressive fibrosis is the underlying pathophysiological process of CKD, and targeted prevention or reversal of the profibrotic cell phenotype is an important goal in developing therapeutics for CKD. Nanoparticles offer new ways to deliver antifibrotic therapies to damaged tissues and resident cells to limit manifestation of the profibrotic phenotype. METHODS: We focused on delivering plasmid DNA expressing bone morphogenetic protein 7 (BMP7) or hepatocyte growth factor (HGF)-NK1 (HGF/NK1) by encapsulation within chitosan nanoparticles coated with hyaluronan, to safely administer multifunctional nanoparticles containing the plasmid DNA to the kidneys for localized and sustained expression of antifibrotic factors. We characterized and evaluated nanoparticles in vitro for biocompatibility and antifibrotic function. To assess antifibrotic activity in vivo, we used noninvasive delivery to unilateral ureteral obstruction mouse models of CKD. RESULTS: Synthesis of hyaluronan-coated chitosan nanoparticles containing plasmid DNA expressing either BMP7 or NGF/NKI resulted in consistently sized nanoparticles, which-following endocytosis driven by CD44+ cells-promoted cellular growth and inhibited fibrotic gene expression in vitro. Intravenous tail injection of these nanoparticles resulted in approximately 40%-45% of gene uptake in kidneys in vivo. The nanoparticles attenuated the development of fibrosis and rescued renal function in unilateral ureteral obstruction mouse models of CKD. Gene delivery of BMP7 reversed the progression of fibrosis and regenerated tubules, whereas delivery of HGF/NK1 halted CKD progression by eliminating collagen fiber deposition. CONCLUSIONS: Nanoparticle delivery of HGF/NK1 conveyed potent antifibrotic and proregenerative effects. Overall, this research provided the proof of concept on which to base future investigations for enhanced targeting and transfection of therapeutic genes to kidney tissues, and an avenue toward treatment of CKD.


Assuntos
Antifibrinolíticos/administração & dosagem , Proteína Morfogenética Óssea 7/genética , Técnicas de Transferência de Genes , Fator de Crescimento de Hepatócito/genética , Nanopartículas Multifuncionais , Insuficiência Renal Crônica/terapia , Animais , Técnicas de Cultura de Células , Quitosana , Modelos Animais de Doenças , Ácido Hialurônico , Camundongos , Polímeros
7.
Acta Biomater ; 113: 289-304, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32663662

RESUMO

Therapeutic angiogenesis with mesenchymal stem cells (MSCs) is promising for the clinical treatment of peripheral artery disease (PAD). However, the heterogeneous proangiogenic nature of MSCs is a key challenge in developing more effective treatments with MSCs for therapeutic angiogenesis purposes. Here, we propose to enhance the therapeutic function of human placenta-derived MSCs (hP-MSCs) in hindlimb ischemia therapy by using nitric oxide (NO)-releasing chitosan hydrogel (CS-NO). Our data showed that the co-transplantation of CS-NO hydrogel with hP-MSCs remarkably improved the grafting of hP-MSCs and ameliorated the functional recovery of ischemic hindlimbs. Moreover, we found that the neovascularization of damaged hindlimbs was significantly increased after co-transplanting CS-NO hydrogel and hP-MSCs, as confirmed by bioluminescence imaging (BLI). Further analysis revealed an endothelial-like status transformation of hP-MSCs in the presence of NO, which was identified as a potential mechanism contributing to the enhanced endothelium-protective and proangiogenic capacities of hP-MSCs that promote angiogenesis in mouse models of hindlimb ischemia. In conclusion, this study provides a promising approach for using NO hydrogel to improve the proangiogenic potency of MSCs in ischemic diseases, and the strategy used here facilitates the development of controlled-release scaffolds for enhancing the therapeutic efficiency of MSCs in angiogenic therapy. STATEMENT OF SIGNIFICANCE: The heterogeneous proangiogenic nature of mesenchymal stem cells (MSCs) is a key challenge in developing more effective treatments with MSCs for therapeutic angiogenesis purposes. In this study, we investigated whether nitric oxide (NO)-releasing chitosan hydrogel (CS-NO) could improve the proangiogenic potency of MSCs in ischemic diseases. Our results revealed an endothelial-like status transformation of human placenta-derived MSCs (hP-MSCs) in the presence of NO, which was identified as a potential mechanism contributing to the enhanced endothelium-protective and proangiogenic capacities of hP-MSCs that promote angiogenesis in mouse models of hindlimb ischemia. The strategy for enhancing the pro-angiogenic activity of MSCs with biomaterials provides a practical idea for overcoming the challenges associated with the clinical application of MSCs in therapeutic angiogenesis.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Feminino , Membro Posterior , Hidrogéis , Isquemia/terapia , Neovascularização Fisiológica , Óxido Nítrico , Gravidez
8.
Adv Sci (Weinh) ; 7(11): 1903516, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32537407

RESUMO

Percutaneous coronary intervention for coronary artery disease treatment often results in pathological vascular injury, characterized by P-selectin overexpression. Adipose-derived stem cells (ADSCs) therapeutic efficacy remains elusive due to poor ADSCs targeting and retention in injured vessels. Here, conjugated P-selectin binding peptide (PBP) to polyethylene glycol-conjugated phospholipid derivative (DMPE-PEG) linkers (DMPE-PEG-PBP; DPP) are used to facilitate the modification of PBP onto ADSCs cell surfaces via hydrophobic interactions between DMPE-PEG and the phospholipid bilayer. DPP modification neither has influence on ADSCs proliferation nor apoptosis/paracrine factor gene expression. A total of 5 × 10-6 m DPP-modified ADSCs (DPP-ADSCs) strongly binds to P-selectin-displaying activated platelets and endothelial cells (ECs) in vitro and to wire-injured rat femoral arteries when administered by intra-arterial injection. Targeted binding of ADSCs shields injury sites from platelet and leukocyte adhesion, thereby decreasing inflammation at injury sites. Furthermore, targeted binding of ADSCs recovers injured ECs functionality and reduces platelet-initiated vascular smooth muscle cells (VSMCs) chemotactic migration. Targeted binding of DPP-human ADSCs to balloon-injured human femoral arteries is also demonstrated in ex vivo experiments. Overall, DPP-ADSCs promote vascular repair, inhibit neointimal hyperplasia, increase endothelium functionality, and maintain normal VSMCs alignment, supporting preclinical noninvasive utilization of DPP-ADSCs for vascular injury.

9.
Nano Lett ; 19(3): 1560-1569, 2019 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-30789273

RESUMO

Bioactive peptides derived from proteins generally need to be folded into secondary structures to activate downstream signaling pathways. However, synthetic peptides typically form random-coils, thus losing their bioactivities. Here, we show that by introducing a self-assembling peptide motif and using different preparation pathways, a peptide from insulin-like growth factor-I (IGF-1) can be folded into an α-helix and ß-sheet. The ß-sheet one exhibits a low dissociation constant to the IGF-1 receptor (IGF-1R, 11.5 nM), which is only about 3 times higher than that of IGF-1 (4.3 nM). However, the α-helical one and the peptide without self-assembling motif show weak affinities to IGF-1R ( KD = 179.1 and 321.6 nM, respectively). At 10 nM, the ß-sheet one efficiently activates the IGF-1 downstream pathway, significantly enhancing HUVEC proliferation and preventing cell apoptosis. The ß-sheet peptide shows superior performance to IGF-1 in vivo, and it improves ischemic hind-limb salvage by significantly reducing muscle degradation and enhancing limb vascularization. Our study provides a useful strategy to constrain peptides into different conformations, which may lead to the development of supramolecular nanomaterials mimicking biofunctional proteins.


Assuntos
Fator de Crescimento Insulin-Like I/química , Nanofibras/química , Peptídeos/química , Receptor IGF Tipo 1/química , Apoptose/genética , Proliferação de Células/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Nanoestruturas/química , Conformação Proteica em alfa-Hélice/efeitos dos fármacos , Conformação Proteica em Folha beta/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Transdução de Sinais/genética
10.
Nat Chem Biol ; 15(2): 151-160, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30598545

RESUMO

The spatiotemporal generation of nitric oxide (NO), a versatile endogenous messenger, is precisely controlled. Despite its therapeutic potential for a wide range of diseases, NO-based therapies are limited clinically due to a lack of effective strategies for precisely delivering NO to a specific site. In the present study, we developed a novel NO delivery system via modification of an enzyme-prodrug pair of galactosidase-galactosyl-NONOate using a 'bump-and-hole' strategy. Precise delivery to targeted tissues was clearly demonstrated by an in vivo near-infrared imaging assay. The therapeutic potential was evaluated in both rat hindlimb ischemia and mouse acute kidney injury models. Targeted delivery of NO clearly enhanced its therapeutic efficacy in tissue repair and function recovery and abolished side effects due to the systemic release of NO. The developed protocol holds broad applicability in the targeted delivery of important gaseous signaling molecules and offers a potent tool for the investigation of relevant molecular mechanisms.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Óxido Nítrico/administração & dosagem , Óxido Nítrico/metabolismo , Animais , Compostos Azo , Galactosidases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Óxido Nítrico/fisiologia , Pró-Fármacos , Ratos , Ratos Sprague-Dawley , beta-Galactosidase/metabolismo , beta-Galactosidase/fisiologia
11.
J Cancer ; 9(24): 4706-4711, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30588255

RESUMO

Purpose: Based on a large-population analysis, we aimed to estimate the incidence and survival of bone metastases (BM) in initial bladder cancer (BC) patients and to identify the risk and prognostic factors of BC patients with BM. Patients and methods: Using the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) database, bladder cancer patients diagnosed between 2010 and 2014 were retrieved. Multivariate logistic and Cox regression analyses were employed to identify risk factors and prognostic factors for BM in BC patients. A Kaplan-Meier analysis with log-rank test was used to estimate the overall survival for BC and the difference between the survival curves. Results: A total of 1,223 (1.39%) BC patients were diagnosed with de novo BM. Variables such as age between 41 to 60 years, black race, unmarried status, higher T stage, higher N stage, poor tumour differentiation grade, lung metastases, liver metastases, and brain metastases were positively associated with BM occurrence. The median survival for BC patients with BM was dramatically decreased to 4.0 months. Factors including advanced age, absence of surgery, and presence of lung, liver, or brain metastases all predicted worse survival. Conclusion: BM can dramatically decrease the survival of bladder cancer patients. The findings of the present study can provide population-based identification of risk and prognostic factors for BC patients with BM at initial diagnosis, which can be used for BM occurrence prediction and individualized treatment plan-making.

12.
Cancer Manag Res ; 10: 2935-2944, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30214288

RESUMO

PURPOSE: The purpose of this study was to assess the incidence of and the risk factors and prognostic factors for bone metastasis (BM) in initial metastatic renal cell carcinoma (RCC) based on a large population analysis. PATIENTS AND METHODS: Data were obtained for a total of 45,824 RCC patients recorded in the database of the Surveillance, Epidemiology, and End Results program of the National Cancer Institute between 2010 and 2014. Multivariate logistic and Cox regression analyses were used to identify the risk factors and prognostic factors associated with BM in RCC patients. Kaplan-Meier analysis was used to estimate the overall survival of RCC patients, and the difference between the survival curves was tested by log-rank tests. RESULTS: A total of 1,509 (3.29%) patients were diagnosed with bone metastases at initial diagnosis. Male gender, higher T stage, lymph node involvement, poor tumor grade, presence of lung, liver, and brain metastases, and the collecting duct type of RCC were positively associated with BM occurrence. The median survival time for RCC patients with bone metastases was 12.0 (95% confidence interval [CI]: 10.69-13.31) months, and the survival time for those with collecting duct, clear-cell, papillary, and chromophobe subtypes of RCC were 3 (95% CI: 0.23-5.77), 13 (95% CI: 11.60-14.40), 8 (95% CI: 5.09-10.91), and 11 (95% CI: 5.02-16.98) months; these differences were significantly different (P<0.01). Older age, higher T stage, lymph node involvement, poor tumor grade, the presence of lung, liver, and brain metastases, collecting duct RCC, and the absence of surgical treatments were the factors associated with worse prognoses. CONCLUSION: BM was highly prevalent and significantly decreased the survival rate of RCC patients. A number of factors associated with the development and prognosis of BM were identified, and these insights provide preventive guidelines for screening and treatment of BM in RCC patients.

13.
Ecotoxicol Environ Saf ; 164: 61-68, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30098506

RESUMO

Antimony, one of the heavier pnictogens, is widely used in industry, and its toxicity has become a major concern. Although previous studies suggested that antimony might be a tumorigenic risk factor in several cancers, the molecular basis underlying antimony-mediated transformation remains unclear. Our results showed that the serum concentration of antimony was higher in prostate cancer specimens relative to that of benign prostate tissues, and this high serum concentration of antimony was closely associated with poorer outcome in prostate cancer patients. Additionally, we demonstrated that antimony could promote prostate cancer cell growth in vitro and in vivo. In order to gain insight into the potential mechanisms, we examined the effects of antimony exposure on downstream signaling that could contribute to tumor development. We found that low-dose antimony could regulate the expression of Ctbp2 by binding and regulating the activity of its MRE domain. Meanwhile, CtBP2 could transcriptionally regulate the expression of RhoC, which is a member of the RhoGTPase family. Subsequently, the kinase activity of ROCK1 is increased, which promotes the stability of oncogene c-Myc. Overall, our study demonstrated that antimony could enhance c-Myc protein stability and promote prostate cancer cell proliferation through activating CtBP2-ROCK1 signaling pathway. These findings also substantially highlighted the potential of targeting molecules within antimony induced CtBP2-c-Myc signaling pathway as a promising therapeutic approach for the treatment of prostate cancer.


Assuntos
Oxirredutases do Álcool/metabolismo , Antimônio/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Quinases Associadas a rho/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Correpressoras , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais
14.
Cancer Manag Res ; 10: 1639-1646, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29970963

RESUMO

PURPOSE: Using the Surveillance, Epidemiology, and End Results database (SEER) to assess the incidence and risk factors of morbidity and prognosis for bone metastases in initial metastatic prostate cancer. PATIENTS AND METHODS: The records of 249,331 prostate cancer patients in the SEER database, diagnosed between 2010 and 2014, were obtained were obtained to investigate the risk factors for developing bone metastasis, and the records of 9925 of them who registered before 2013 were retrieved (with at least 1 year follow up) to explore the prognostic factors for bone metastasis. Multivariate logistic and Cox regression were used to identify risk factors and prognostic factors for bone metastases, respectively. RESULTS: In total, 12,794 patients (5.1%) were diagnosed with bone metastases at the initial diagnosis. Older age, unmarried status, lymph node metastasis, poor tumor differentiation grade (Gleason grade), metastases at lung, brain, and liver were all positively associated with risk for the morbidity and prognosis of bone metastasis in prostate cancer. Black race and higher T stage were positively associated with bone metastasis development; however, they were not associated with a prognosis of bone metastasis. CONCLUSION: The incidence of bone metastasis in prostate cancer was approximately 5% with poor survival. The prostate cancer has homogeneous and heterogeneous risk factors for incidence and prognosis of bone metastasis, which may provide potential guidelines for the screening and preventive treatment for the bone metastasis of prostate cancer.

15.
Toxicol Lett ; 288: 136-142, 2018 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-29462692

RESUMO

Antimony is a widely used heavier pnictogens in industry, and its toxicity has been a matter of concern. Although previous studies have suggested that antimony may have the function as either a tumor suppressor or an oncogene in several cancers, the molecular basis underlying antimony-mediated transformation is still unclear. In the current study, we attempt to elucidate the potential role of antimony in the development of prostate cancer. Our results showed that the concentration of antimony was much higher in serum of prostate cancer patients, and was closely associated with poor outcome of patients who underwent radical prostatectomy. Additionally, low dose of antimony could promote proliferation and invasion of androgen-dependent prostate cancer cell line LNCaP cells in vitro and in vivo. The mechanistic studies demonstrated that exposure to antimony triggered the phosphorylation of androgen receptor (AR), which transcriptionally regulates the expression of androgen-related targets, including PSA and NKX3.1. Overall, our results unearthed that antimony could promote tumor growth by mimicking androgen activity in androgen-dependent prostate cancer cells. Therefore, these findings expanded our understanding on the molecular mechanism of antimony in tumorigenesis and tumor progression of prostate cancer, and it appears to be an inspiring strategy to restrain prostate cancer by inhibiting antimony-induced androgen-like effects.


Assuntos
Androgênios/farmacologia , Antimônio/farmacologia , Neoplasias da Próstata/patologia , Antagonistas de Androgênios/farmacologia , Animais , Antimônio/sangue , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Antígeno Prostático Específico/biossíntese , Antígeno Prostático Específico/efeitos dos fármacos , Prostatectomia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/cirurgia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Androgênicos/efeitos dos fármacos , Fatores de Transcrição/biossíntese , Fatores de Transcrição/efeitos dos fármacos , Resultado do Tratamento
16.
ACS Appl Mater Interfaces ; 10(5): 4481-4493, 2018 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-29327586

RESUMO

Stem cell treatment for critical limb ischemia yields a limited therapeutic effect due to cell loss and dysfunction caused by local ischemic environment. Biomimetic scaffolds emerge as ideal cell delivery vehicles for regulating cell fate via mimicking the components of stem cell niche. Herein, we prepared a bioactive hydrogel by mixing chitosan and hyaluronic acid that is immobilized with C domain peptide of insulin-like growth factor 1 (IGF-1C) and examined whether this hydrogel could augment stem cell survival and therapeutic potential. Our results showed that IGF-1C-modified hydrogel increased in vitro viability and proangiogenic activity of adipose-derived stromal cells (ADSCs). Moreover, cotransplantation of hydrogel and ADSCs into ischemic hind limbs of mice effectively ameliorated blood perfusion and muscle regeneration, leading to superior limb salvage. These therapeutic effects can be ascribed to improved ADSC retention, angiopoientin-1 secretion, and neovascularization, as well as reduced inflammatory cell infiltration. Additionally, hydrogel enhanced antifibrotic activity of ADSCs, as evidenced by decreased collagen accumulation at late stage. Together, our findings indicate that composite hydrogel modified by IGF-1C could promote survival and proangiogenic capacity of ADSCs and thereby represents a feasible option for cell-based treatment for critical limb ischemia.


Assuntos
Transplante de Células-Tronco , Tecido Adiposo , Animais , Células Cultivadas , Extremidades , Hidrogéis , Isquemia , Camundongos , Neovascularização Fisiológica
17.
Oncotarget ; 8(50): 87390-87400, 2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-29152089

RESUMO

Mesangial cells (MCs), which are vascular smooth muscle-derived cells, occupy the central position in the glomerulus. Diabetic nephropathy (DN) is one of the most common diabetes complications and is likely attributed to the loss of MC contractility. Myocardin stimulates downstream vascular smooth muscle genes and regulates the contractility of vascular smooth muscle cells. Therefore, we hypothesized that myocardin is expressed in MCs and that high glucose is involved in the regulation of myocardin and downstream contractile genes in the context of DN. Confocal microscopy revealed that myocardin is expressed in rat MCs. Western blot and RT-qPCR analyses showed that treatment with 30 mM D-glucose significantly downregulated the mRNA and protein levels of myocardin and downstream SM α-actin. As an isotonic contrast, 30 mM mannitol did not affect myocardin mRNA levels but did downregulate myocardin protein levels. Treatment with 30 mM mannitol also downregulated SM α-actin mRNA and protein levels. Conversely, as another isotonic contrast, 30 mM L-glucose also had no effect on myocardin and SM α-actin expression in MCs. The extracellular signal-regulated kinase (ERK) pathway was activated by treatment with 30 mM D-glucose or mannitol, while specific inhibitors of the ERK pathway (PD98059) compromised the downregulation of myocardin and SM α-actin triggered by high glucose or mannitol. Thus we revealed that myocardin is expressed in MCs and that high glucose downregulates myocardin expression and downstream contractile protein SM α-actin via the ERK pathway. Our results suggest a novel mechanism for high glucose inhibition of MC contraction, which contributes to DN pathogenesis.

18.
Curr Med Chem ; 23(24): 2579-2601, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27480214

RESUMO

Nitric oxide (NO), as an essential signaling molecule, participates in various physiological processes such as cardiovascular homeostasis, neuronal transmission, immunomodulation, and tumor growth. The multiple role of NO in physiology and pathophysiology has triggered a massive interest in the strategies of delivering exogenous NO for biomedical applications. Hence, different kinds of NO prodrugs have been developed up to date, including diazeniumdiolates, S-nitrosothiol, metal-nitrosyl, nitrobenzene, and so on. However, the clinical application of these low molecular weight NO donors has been restricted due to the problems of burst release, low payloads, and untargeted delivery. The delivery of NO by biomaterialbased carrier offers a beneficial strategy to realize the controlled and sustained delivery of NO to the targeted tissues or organs. In detail, NO-donor prodrugs have been attached and loaded to diverse biomaterials to fabricate nanoparticles, hydrogels, and coating platforms by means of physical, chemical, or supramolecular techniques. These NO-releasing biomaterials hold promise for a number of biomedical applications ranging from therapy of the ischemic disease and several types of cancer to cardiovascular devices and wound dressing. First, surface coating with NO-releasing biomaterials could mimic the physiological function of vascular endothelium, therefore promoting vascularization and improving the patency of cardiovascular implants. Next, because NO also mediates many important processes that take place after cutaneous injury, NO-releasing biomaterials could serve as ideal wound dressing to accelerate tissue regeneration. Finally, biomaterials enable localized delivery of high dose of NO to tumors in a sustained manner, thus generating potent tumoricidal effect. In this review, we will summarize the progress of different NO-releasing biomaterials, and highlight their biomedical applications with a hope to inspire new perspectives in the area of biomaterial-based NO-delivery systems.


Assuntos
Materiais Biocompatíveis/química , Portadores de Fármacos/química , Óxido Nítrico/metabolismo , Animais , Doenças Cardiovasculares/tratamento farmacológico , Humanos , Hidrogéis/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Óxido Nítrico/química , Doadores de Óxido Nítrico/administração & dosagem , Doadores de Óxido Nítrico/química , Polímeros/química , Stents
19.
Stem Cell Res Ther ; 7(1): 95, 2016 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-27460364

RESUMO

BACKGROUND: Microenvironment is being increasingly recognized as a critical determinant in tumor progression and metastasis. However, the appropriate regulatory mechanism to maintain the normal balance between differentiation and self-renewal of the cancer cell in microenvironment is not well known. METHODS: 4T1 breast cancer cells were treated with embryonic stem (ES) cell conditioned medium which was collected from mouse ES cells. Inhibition of tumor cell growth was based on the reduction of cell proliferation and viability, and inhibition of aggressive properties of tumor cells were examined using the wound-healing and mammosphere assays. The expression of stem cell-associated genes was detected by quantitative RT-PCR. RESULTS: We used a real-time imaging system to investigate the effect of the mouse ES cell microenvironment on aggressive breast cancer cells in vitro and in vivo. Exposure of breast cancer cells in mouse ES cell conditioned medium resulted in inhibition of growth, migration, metastasis, and angiogenesis of cancer cells. For many tumors, aggressive properties were tightly related to Stat3 signaling activation. We specifically discovered that the ES cell microenvironment sufficiently suppressed Stat3 signaling pathway activation in aggressive tumor cells, leading to a reduction in tumorigenesis and invasiveness. CONCLUSIONS: We identified important functions of Stat3 and their implications for antitumor effects of ES cell conditioned medium. Some factors secreted by ES cells could efficiently suppress Stat3 pathway activation in breast cancer cells, and were then involved in cancer cell growth, survival, invasion, and migration. This study may act as a platform to understand tumor cell plasticity and may offer new therapeutic strategies to inhibit breast cancer progression.


Assuntos
Carcinogênese/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Embrionárias/metabolismo , Neoplasias Mamárias Animais/tratamento farmacológico , Nicho de Células-Tronco/genética , Animais , Bioensaio , Carcinogênese/genética , Carcinogênese/metabolismo , Carcinogênese/patologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células Alimentadoras/citologia , Células Alimentadoras/metabolismo , Feminino , Expressão Gênica , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Cicatrização/efeitos dos fármacos
20.
Adv Healthc Mater ; 5(18): 2376-85, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27391702

RESUMO

The lack of efficient vascularization within frequently used poly(ε-caprolactone) (PCL) scaffolds has hindered their application in tissue engineering. Hydrophobin HGFI, an amphiphilic protein, can form a self-assembly layer on the surface of PCL scaffolds and convert their wettability. In this study, a fusion protein consisting of HGFI and vascular endothelial growth factor (VEGF) is prepared by Pichia pastoris expression system. Sodium dodecyl sulface-polyacrylamide gel electrophoresis (SDS-PAGE) and western blotting confirm that the VEGF-HGFI is successfully isolated and purified. Transmission electron microscope and water contact angle measurement demonstrate that VEGF-HGFI can form a self-assembly layer with about 25 nm in thickness on electrospun PCL fibers and increase their hydrophilicity. VEGF-HGFI modification can effectively enhance the adhesion, migration, and proliferation of human umbilical vein endothelial cells. Near-infrared fluorescence imaging shows that the VEGF-HGFI modification on PCL scaffolds can exist at least 21 d in vitro and at least 14 d in vivo. Bioluminescence imaging shows that VEGF-HGFI can effectively activate vascular endothelial growth factor receptor 2 receptors. Subcutaneous implantation in mice and rats reveal that cellularization and vascularization are significantly improved in VEGF-HGFI modified PCL scaffolds. These results suggest that VEGF-HGFI is a useful molecule for functional modification of scaffolds to enhance cellularization and vascularization in tissue engineering.


Assuntos
Materiais Revestidos Biocompatíveis/química , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neovascularização Fisiológica , Poliésteres/química , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/química , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Proteínas Recombinantes de Fusão/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...