Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Sci ; 113(2): 733-743, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34859546

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Hidrogeles/uso terapéutico , Mifepristona/uso terapéutico , Paclitaxel/uso terapéutico , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Disponibilidad Biológica , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Portadores de Fármacos/uso terapéutico , Liberación de Fármacos , Femenino , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Ratones , Mifepristona/química , Mifepristona/farmacología , Nanoestructuras/uso terapéutico , Paclitaxel/química , Paclitaxel/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Nat Chem Biol ; 15(2): 151-160, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30598545

RESUMEN

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.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Animales , Compuestos Azo , Galactosidasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Óxido Nítrico/fisiología , Profármacos , Ratas , Ratas Sprague-Dawley , beta-Galactosidasa/metabolismo , beta-Galactosidasa/fisiología
3.
J Am Soc Nephrol ; 31(10): 2292-2311, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32769144

RESUMEN

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.


Asunto(s)
Antifibrinolíticos/administración & dosificación , Proteína Morfogenética Ósea 7/genética , Técnicas de Transferencia de Gen , Factor de Crecimiento de Hepatocito/genética , Nanopartículas Multifuncionales , Insuficiencia Renal Crónica/terapia , Animales , Técnicas de Cultivo de Célula , Quitosano , Modelos Animales de Enfermedad , Ácido Hialurónico , Ratones , Polímeros
4.
Nano Lett ; 19(3): 1560-1569, 2019 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-30789273

RESUMEN

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.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/química , Nanofibras/química , Péptidos/química , Receptor IGF Tipo 1/química , Apoptosis/genética , Proliferación Celular/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Nanoestructuras/química , Conformación Proteica en Hélice alfa/efectos de los fármacos , Conformación Proteica en Lámina beta/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Transducción de Señal/genética
5.
Ecotoxicol Environ Saf ; 164: 61-68, 2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30098506

RESUMEN

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.


Asunto(s)
Oxidorreductasas de Alcohol/metabolismo , Antimonio/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Proteínas Co-Represoras , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal
6.
J Am Soc Nephrol ; 27(8): 2357-69, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26869006

RESUMEN

Low cell retention and engraftment after transplantation limit the successful application of stem cell therapy for AKI. Engineered microenvironments consisting of a hydrogel matrix and growth factors have been increasingly successful in controlling stem cell fate by mimicking native stem cell niche components. Here, we synthesized a bioactive hydrogel by immobilizing the C domain peptide of IGF-1 (IGF-1C) on chitosan, and we hypothesized that this hydrogel could provide a favorable niche for adipose-derived mesenchymal stem cells (ADSCs) and thereby enhance cell survival in an AKI model. In vitro studies demonstrated that compared with no hydrogel or chitosan hydrogel only, the chitosan-IGF-1C hydrogel increased cell viability through paracrine effects. In vivo, cotransplantation of the chitosan-IGF-1C hydrogel and ADSCs in ischemic kidneys ameliorated renal function, likely by the observed promotion of stem cell survival and angiogenesis, as visualized by bioluminescence imaging and attenuation of fibrosis. In conclusion, IGF-1C immobilized on a chitosan hydrogel provides an artificial microenvironment for ADSCs and may be a promising therapeutic approach for AKI.


Asunto(s)
Lesión Renal Aguda/terapia , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Trasplante de Células Madre Mesenquimatosas , Tejido Adiposo/citología , Animales , Quitosano , Terapia Combinada , Hidrogel de Polietilenoglicol-Dimetacrilato , Ratones
7.
Hum Reprod ; 30(1): 61-70, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25398968

RESUMEN

STUDY QUESTION: Does tumor necrosis factor-α (TNF-α) differentially regulate matrix metalloproteinase-2 (MMP-2) expression in leiomyomas compared with normal myometrium? SUMMARY ANSWER: TNF-α up-regulates MMP-2 expression and stimulates cell migration through the activation of extracellular signal-regulated kinase (ERK) signaling pathway in leiomyoma smooth muscle cells (SMCs), but not in normal myometrial SMCs. WHAT IS KNOWN ALREADY: Uterine leiomyoma, the benign smooth muscle cell tumor, is the single most common indication for hysterectomy. High expression of MMPs or TNF-α has been reported in uterine leiomyomas; however, the molecular mechanism underlying these observations remains unknown. STUDY DESIGN, SIZE, DURATION: Samples were obtained between 2009 and 2013 from 12 women of reproductive age at the proliferative phase of the menstrual cycle by hysterectomy. Leiomyomas and matched normal myometrium from each woman were analyzed in vitro. PARTICIPANTS/MATERIALS, SETTING, METHODS: Western blot, RT-qPCR and a wound-healing assay were used to investigate the effects of TNF-α on MMP-2 expression and intracellular signal transduction in cultured SMCs from leiomyomas and matched myometrium. MAIN RESULTS AND THE ROLE OF CHANCE: Western blot and RT-qPCR analyses using tissues from clinical patients showed that the levels of MMP-2 protein (P = 0.008) and mRNA (P = 0.009) were significantly higher in uterine leiomyomas compared with their matched myometrium. Treatment with TNF-α significantly up-regulated the protein (P = 0.039) and mRNA (P = 0.037) levels of MMP-2 in cultured leiomyoma SMCs but not in matched myometrial SMCs. The extracellular signal-regulated kinase (ERK) and nuclear factor-kappa B (NF-κB) pathways were activated by TNF-α in leiomyoma SMCs. Specific inhibitors of the ERK or NF-κB pathway (PD98059 or Bay11-7082) suppressed TNF-α-induced MMP-2 expression in leiomyoma SMCs. The wound-healing assay revealed that TNF-α promoted the migration of cultured leiomyoma SMCs (P = 0.036); however, PD98059 compromised the cell migration triggered by TNF-α. LIMITATIONS, REASONS FOR CAUTION: This study is descriptive and although we observed clear differential regulation of MMP-2 by TNF-α at mRNA and protein levels in leiomyoma, future studies are needed to identify why the difference in TNF-α response exists between human leiomyoma tissue and normal myometrium. Including some of the experiments such as transfection studies for TNF-α and MMP-2 promoter mapping could have added more insight as to why this difference exists. In addition, further studies in vivo are needed to verify the results obtained from primary cultured SMCs. WIDER IMPLICATIONS OF THE FINDINGS: Considering the positive effect of TNF-α on leiomyoma SMC migration, strategies targeting TNF-α, in parallel with the production of more specific inhibitors of MMPs, may provide alternative therapeutic approaches for the treatment of leiomyoma. STUDY FUNDING/COMPETING INTERESTS: This work was partially supported by grants from the Program for New Century Excellent Talents in University (NCET-12-0282), National Natural Science Foundation of China (81371620) and Tianjin Natural Science Foundation (12JCZDJC24900). The authors have no conflicts of interest to declare.


Asunto(s)
Leiomioma/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Miometrio/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Adulto , Movimiento Celular , Células Cultivadas , Femenino , Fase Folicular/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Fa Yi Xue Za Zhi ; 31(4): 262-5, 2015 Aug.
Artículo en Zh | MEDLINE | ID: mdl-26665877

RESUMEN

OBJECTIVE: To observe cardiac ultrastructure and the expression of heat shock protein 70 (HSP70) and hypoxia inducible factor-lα (HIF-lα) in electric shock death rats and to explore the application of these indexes as the basis of medical identification in electric shock death. METHODS: Seventy-two SD rats were randomly divided into electric shock death group, postmortem electric shock group and the control group. The changes of myocardial ultrastructure were observed by transmission electron microscope, and the expressions of myocardial HSP70 and HIF-1α were observed by immunohistochemical technology. RESULTS: Myocardial myofibril fracture, mitochondrial cristae and membrane dissolution, and disordered arrangement of Z lines and M lines were observed in electric shock rats. HSP70 and HIF-lα were strong positive expressions in the electric shock death group, significantly compared with the control and postmortem electric shock groups (P < 0.05). CONCLUSION: The expressions of HSP70 and HIF-lα were obviously increased in electric shock death group, which may be used as the diagnostic indicator of electric shock death.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocardio/metabolismo , Miocardio/patología , Animales , Muerte , Ratas , Ratas Sprague-Dawley
9.
Cell Physiol Biochem ; 32(5): 1517-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24335177

RESUMEN

BACKGROUND: Despite increasing attention on the role of bone marrow derived stem cells in repair or rejuvenation of tissues and organs, cellular mechanisms of such cell-based therapy remain poorly understood. METHODS: We reconstituted hematopoiesis in recipient C57BL/6J mice by transplanting syngeneic GFP(+) bone marrow (BM) cells. Subsequently, the recipients received subcutaneous injection of granulocyte-colony stimulating factor (G-CSF) and were subjected to acute renal ischemic injury. Flow cytometry and immunostaining were performed at various time points to assess engraftment and phenotype of BM derived stem cells. RESULTS: Administration of G-CSF increased the release of BM derived stem cells into circulation and enhanced the ensuing recruitment of BM derived stem cells into injured kidney. During the second month post injury, migrated BM derived stem cells lost hematopoietic phenotype (CD45) but maintained the expression of other markers (Sca-1, CD133 and CD44), suggesting their potential of transdifferentiation into renal stem cells. Moreover, G-CSF treatment enhanced the phenotypic conversion. CONCLUSION: Our work depicted a time-course dependent transition of phenotypic characteristics of BM derived stem cells, demonstrated the existence of BM derived stem cells in damaged kidney and revealed the effects of G-CSF on cell transdifferentiation.


Asunto(s)
Lesión Renal Aguda/patología , Células Madre Hematopoyéticas/fisiología , Animales , Trasplante de Médula Ósea , Transdiferenciación Celular , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/metabolismo , Riñón/irrigación sanguínea , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Daño por Reperfusión/patología , Factores de Tiempo
10.
Cancer Med ; 12(13): 14149-14156, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37325945

RESUMEN

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.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Estudios de Cohortes , Pronóstico , Estudios Retrospectivos , Inhibidores de Proteínas Quinasas/uso terapéutico
11.
BMC Nephrol ; 13: 105, 2012 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22963129

RESUMEN

BACKGROUND: Bone marrow (BM) stem cells have been reported to contribute to tissue repair after kidney injury model. However, there is no direct evidence so far that BM cells can trans-differentiate into renal stem cells. METHODS: To investigate whether BM stem cells contribute to repopulate the renal stem cell pool, we transplanted BM cells from transgenic mice, expressing enhanced green fluorescent protein (EGFP) into wild-type irradiated recipients. Following hematological reconstitution and ischemia-reperfusion (I/R), Sca-1 and c-Kit positive renal stem cells in kidney were evaluated by immunostaining and flow cytometry analysis. Moreover, granulocyte colony stimulating factor (G-CSF) was administrated to further explore if G-CSF can mobilize BM cells and enhance trans-differentiation efficiency of BM cells into renal stem cells. RESULTS: BM-derived cells can contribute to the Sca-1(+) or c-Kit(+) renal progenitor cells population, although most renal stem cells came from indigenous cells. Furthermore, G-CSF administration nearly doubled the frequency of Sca-1+ BM-derived renal stem cells and increased capillary density of I/R injured kidneys. CONCLUSIONS: These findings indicate that BM derived stem cells can give rise to cells that share properties of renal resident stem cell. Moreover, G-CSF mobilization can enhance this effect.


Asunto(s)
Lesión Renal Aguda/patología , Lesión Renal Aguda/cirugía , Células de la Médula Ósea/patología , Trasplante de Médula Ósea/métodos , Daño por Reperfusión/patología , Daño por Reperfusión/cirugía , Células Madre/patología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Células Madre/métodos , Resultado del Tratamiento
12.
J Oncol ; 2021: 5572402, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34539784

RESUMEN

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.

13.
Biomed Res Int ; 2021: 8249293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805404

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Cinesinas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Biología Computacional , Progresión de la Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Estudios Retrospectivos , Regulación hacia Arriba
14.
Acta Biomater ; 128: 209-221, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33878473

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Andamios del Tejido , Animales , Matriz Extracelular , Isquemia/terapia , Ratones , Ratas , Trasplante de Células Madre , Ingeniería de Tejidos
15.
Acta Biomater ; 113: 289-304, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32663662

RESUMEN

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.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Femenino , Miembro Posterior , Hidrogeles , Isquemia/terapia , Neovascularización Fisiológica , Óxido Nítrico , Embarazo
16.
Adv Sci (Weinh) ; 7(11): 1903516, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32537407

RESUMEN

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.

17.
Toxicol Lett ; 288: 136-142, 2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29462692

RESUMEN

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.


Asunto(s)
Andrógenos/farmacología , Antimonio/farmacología , Neoplasias de la Próstata/patología , Antagonistas de Andrógenos/farmacología , Animales , Antimonio/sangre , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Fosforilación , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/efectos de los fármacos , Prostatectomía , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/cirugía , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Androgénicos/efectos de los fármacos , Factores de Transcripción/biosíntesis , Factores de Transcripción/efectos de los fármacos , Resultado del Tratamiento
18.
Cancer Manag Res ; 10: 2935-2944, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30214288

RESUMEN

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.

19.
Cancer Manag Res ; 10: 1639-1646, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29970963

RESUMEN

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.

20.
J Cancer ; 9(24): 4706-4711, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30588255

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA