Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carbohydr Polym ; 320: 121238, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37659799

RESUMEN

The healing of wounds in diabetic patients is a huge challenge issue in clinical medicine due to the disordered immune. Recruiting endogenous cells to play a role in the early stage and timely reducing inflammation to promote healing in the middle or late of injuring are both prerequisites for effective treatment. Here, inspired by natural extracellular matrix, three-dimensional porous polyurethane-hyaluronic acid hybrid hydrogel scaffolds (PUHA) were prepared to repair diabetic wound through activate cell immunity by moderate foreign body reaction, provide cell adhesion growth extracellular matrix of hyaluronic acid (HA) and exhibit anti-inflammatory effect of polyurethane (PU). The interaction between PU and HA alters the compact PU hydrogel into macroporous PUHA hydrogel scaffolds with super-swelling, elastic mechanical properties, and controllable degradation, which are suitable for endogenous cells infiltration, growth and immune activation. Additionally, incorporating with RGD, PUHA hydrogel scaffolds with bioactive physicochemical features can evidently reduce the inflammation and modulate the polarization of macrophage apparently both in vitro and in vivo, mainly through downregulation of cytokine-cytokine receptor interaction genes, leading to reprogramming immune-microenvironment and rapid diabetic wound healing. This method of gathering cells initially and intervening immune-microenvironment in time provides an expected way to design biomaterials for chronic wound healing.


Asunto(s)
Diabetes Mellitus , Ácido Hialurónico , Humanos , Ácido Hialurónico/farmacología , Poliuretanos , Hidrogeles/farmacología , Inflamación , Materiales Biocompatibles
2.
Stem Cell Res Ther ; 14(1): 184, 2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37501214

RESUMEN

BACKGROUND: Decompensated liver cirrhosis (DLC), a terminal-stage complication of liver disease, is a major cause of morbidity and mortality in patients with hepatopathies. Human umbilical cord mesenchymal stem cell (hUCMSC) therapy has emerged as a novel treatment alternative for the treatment of DLC. However, optimized therapy protocols and the associated mechanisms are not entirely understood. METHODS: We constructed a DLC rat model consistent with the typical clinical characteristics combined use of PB and CCL4. Performing dynamic detection of liver morphology and function in rats for 11 weeks, various disease characteristics of DLC and the therapeutic effect of hUCMSCs on DLC in experimental rats were thoroughly investigated, according to ascites examination, histopathological, and related blood biochemical analyses. Flow cytometry analysis of rat liver, immunofluorescence, and RT-qPCR was performed to examine the changes in the liver immune microenvironment after hucMSCs treatment. We performed RNA-seq analysis of liver and primary macrophages and hUCMSCs co-culture system in vitro to explore possible signaling pathways. PPARγ antagonist, GW9662, and clodronate liposomes were used to inhibit PPAR activation and pre-exhaustion of macrophages in DLC rats' livers, respectively. RESULTS: We found that changing the two key issues, the frequency and initial phase of hUCMSCs infusion, can affect the efficacy of hUCMSCs, and the optimal hUCMSCs treatment schedule is once every week for three weeks at the early stage of DLC progression, providing the best therapeutic effect in reducing mortality and ascites, and improving liver function in DLC rats. hUCMSCs treatment skewed the macrophage phenotype from M1-type to M2-type by activating the PPARγ signaling pathway in the liver, which was approved by primary macrophages and hUCMSCs co-culture system in vitro. Both inhibition of PPARγ activation with GW9662 and pre-exhaustion of macrophages in DLC rats' liver abolished the regulation of hUCMSCs on macrophage polarization, thus attenuating the beneficial effect of hUCMSCs treatment in DLC rats. CONCLUSIONS: These data demonstrated that the optimal hUCMSCs treatment effectively inhibits the ascites formation, prolongs survival and significantly improves liver structure and function in DLC rats through the activation of the PPARγ signaling pathway within liver macrophages. Our study compared the efficacy of different hUCMSCs infusion regimens for DLC, providing new insights on cell-based therapies for regenerative medicine.


Asunto(s)
Ascitis , PPAR gamma , Ratas , Humanos , Animales , PPAR gamma/genética , Ascitis/terapia , Cirrosis Hepática/terapia , Macrófagos , Cordón Umbilical
3.
Aging (Albany NY) ; 15(11): 4986-5006, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37280069

RESUMEN

The present study aims to construct a predictive model for prognosis and immunotherapy response in lung adenocarcinoma (LUAD). Transcriptome data were extracted from the Cancer Genome Atlas (TCGA), GSE41271, and IMvigor210. The weighted gene correlation network analysis was utilized to identify the hub modules related to immune/stromal cells. Then, univariate, LASSO, and multivariate Cox regression analyses were employed to develop a predictive signature based on genes of the hub module. Moreover, the association between the predictive signature and immunotherapy response was also investigated. As a result, seven genes (FGF10, SERINE2, LSAMP, STXBP5, PDE5A, GLI2, FRMD6) were screened to develop the cancer associated fibroblasts (CAFs)-related risk signature (CAFRS). LUAD patients with high-risk score underwent shortened Overall survival (OS). A strong correlation was found between CAFRS and immune infiltrations/functions. The gene set variation analysis showed that G2/M checkpoint, epithelial-mesenchymal transition, hypoxia, glycolysis, and PI3K-Akt-mTOR pathways were greatly enriched in the high-risk subgroup. Moreover, patients with higher risk score were less likely to respond to immunotherapy. A nomogram based on CAFRS and Stage presented a stronger predictive performance for OS than the single indicator. In conclusion, the CAFRS exhibited a potent predictive value for OS and immunotherapy response in LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Fibroblastos Asociados al Cáncer , Neoplasias Pulmonares , Humanos , Fosfatidilinositol 3-Quinasas , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/terapia , Inmunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Pronóstico
4.
Oncoimmunology ; 11(1): 2127282, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36185809

RESUMEN

A major challenge in natural killer (NK) cell immunotherapy is the limited persistence of NK cells in vivo. However, the proliferation of NK cells is dependent on cytokines such as interleukin-2 (IL-2). Although IL-2 is a critical cytokine for NK cell activation and survival, IL-2 administration in adoptive NK cell therapy can induce adverse toxicities. To improve the persistence of NK cells and attenuate the systemic toxicity of IL-2, we constructed a cell-restricted artificial IL-2, named membrane-bound IL-2 (mbIL-2), comprising human IL-2 and human IL-2Rα joined by a classic linker. We found that mbIL-2-activated NK-92 cells can survive and proliferate in vitro and in vivo, independent of exogenous IL-2, while mbIL-2-expressing NK-92 cells do not support bystander cell survival or proliferation. Additionally, mbIL-2 enhanced NK-92 cell-mediated antitumor activity by tuning the IL-2 receptor downstream signals and NK cell receptor repertoire expression. To conclude, our novel mbIL-2 improves NK-92 cell persistence and enhances NK-92 cell-mediated antitumor activity. NK-92 cells genetically modified to express the novel mbIL-2 with potential significance for clinical development.


Asunto(s)
Interleucina-2 , Células Asesinas Naturales , Citocinas/metabolismo , Humanos , Interleucina-2/metabolismo , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Receptores de Interleucina-2/metabolismo , Receptores de Células Asesinas Naturales/metabolismo
5.
Stem Cell Res Ther ; 13(1): 465, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36076306

RESUMEN

BACKGROUND: Inflammatory bowel diseases (IBD) are chronic relapsing-remitting inflammatory diseases of the gastrointestinal tract that are typically categorized into two subtypes: Crohn's disease (CD) and ulcerative colitis (UC). Although MSCs therapy has achieved encouraging outcomes in IBD therapy, objective responses are limited in colon fibrosis stenosis owing to the complicated microenvironment of CD and MSCs heterogeneity of quality. Here, we chose IFN-γ and kynurenic acid (KYNA) to overcome the low response and heterogeneity of human adipose-derived MSCs (hADSCs) to treat IBD and expand the therapeutic effects based on the excellent ability of IFN-γ and KYNA to promote indoleamine 2,3-dioxygenase-1 (IDO-1) signaling, providing a potential protocol to treat IBD and fibrosis disease. METHODS: hADSCs were isolated, cultured, and identified from human abdominal adipose tissue. The CD pathology-like acute colitis and chronic colon fibrosis rat model was induced by 2,4,6-trinitrobenzen sulfonic acid (TNBS). hADSCs were pretreated in vitro with IFN-γ and KYNA and then were transplanted intravenously at day 1 and 3 of TNBS administration in colitis along with at day 1, 15, and 29 of TNBS administration in chronic colonic fibrosis. Therapeutic efficacy was evaluated by body weights, disease activity index, pathological staining, real-time PCR, Western blot, and flow cytometry. For knockout of IDO-1, hADSCs were transfected with IDO-1-targeting small gRNA carried on a CRISPR-Cas9-lentivirus vector. RESULTS: hADSCs treated with IFN-γ and KYNA significantly upregulated the expression and secretion of IDO-1, which has effectively ameliorated CD pathology-like colitis injury and fibrosis. Notably, the ability of hADSCs with IDO-1 knockout to treat colitis was significantly impaired and diminished the protective effects of the primed hADSCs with IFN-γ and KYNA. CONCLUSION: Inflammatory cytokines IFN-γ- and KYNA-treated hADSCs more effectively alleviate TNBS-induced colitis and colonic fibrosis through an IDO-1-dependent manner. Primed hADSCs are a promising new strategy to improve the therapeutic efficacy of MSCs and worth further research.


Asunto(s)
Colitis , Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Células Madre Mesenquimatosas , Animales , Colitis/inducido químicamente , Enfermedad de Crohn/patología , Fibrosis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Interferón gamma/genética , Interferón gamma/metabolismo , Ácido Quinurénico/efectos adversos , Ácido Quinurénico/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratas
6.
Artículo en Inglés | MEDLINE | ID: mdl-35544723

RESUMEN

Transplantation of encapsulated islets has been shown to hold a promising potential treatment for type 1 diabetes (T1D). However, there are several obstacles to overcome, such as immune rejection by the host of the grafts, sustainability of islet function, and retrievability or replacement of the encapsulated system, hinder their clinical applications. In this study, mini-capsule devices containing islets were fabricated by using digital light processing (DLP) 3D printing. To ensure a high survival rate and low immunogenicity of the fabricated islets, 20s was selected as the most suitable printing condition. Meanwhile, the mini-capsule devices with a groove structure were fabricated to prevent islet cells leakage. Subcutaneous transplantations of encapsulated islets in immunocompetent C57BL/6 mice indicated significant improvement in the symptoms of streptozotocin-induced hyperglycemia without any immunosuppression treatment for at least 15 weeks. In vivo intraperitoneal glucose tolerance tests (IPGTT) performed at different time points demonstrated therapeutically relevant glycemic ameliorate of the device. The implants retrieved after 15 weeks still contained viable and adequate numbers of islet cells. The results of this study indicate that the proposed mini-capsule device can deliver sufficient islet cell mass, prevent islet cells leakage, and maintain long-term cell survival while allowing easy retrieval. Furthermore, the proposed encapsulated islets may help with T1D cellular treatment by overcoming the obstacles of islet transplantation.

7.
Oncogene ; 40(35): 5367-5378, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34272474

RESUMEN

Dexamethasone (Dex), as a pretreatment agent, is widely used to attenuate the side effects of chemotherapy in breast cancer treatment. However, whether and how Dex affects breast cancer metastasis remain to be furtherly understood. In this study, we established several mouse breast cancer metastatic models to study the effect of Dex in vitro and in vivo. Transwell, Western Blot and RNA interference were applied to study the molecular mechanism of Dex in promoting breast cancer cell migration. Meanwhile, the effect of Dex on lung metastasis of breast cancer in Dex combined with PTX chemotherapy was discussed. Our results confirmed that Dex could promote breast cancer cell metastasis both in vitro and in vivo. Mechanistic studies revealed that this pro-metastatic effect of Dex was mediated by the GR-PI3K-SGK1-CTGF pathway in tumor cells. Ligation of Dex and glucocorticoid receptor (GR) on tumor cells activated the PI3K signaling pathway and upregulated serum glucocorticoid-inducible kinase 1 (SGK1) expression, and then increased the expression of connective tissue growth factor (CTGF) through Nedd4l-Smad2. Moreover, Dex was the leading factor for lung metastasis in a standard regimen for breast cancer treatment with paclitaxel and Dex. Importantly, targeting SGK1 with the inhibitor GSK650394 remarkably reduced lung metastasis in this regimen. Our present data provide new insights into Dex-induced breast cancer metastasis and indicate that SGK1 could be a candidate target for the treatment of breast cancer metastasis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Animales , Glucocorticoides , Humanos , Ratones , Fosfatidilinositol 3-Quinasas , Proteínas Serina-Treonina Quinasas
8.
Stem Cell Res Ther ; 12(1): 375, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215318

RESUMEN

BACKGROUND: Direct reprogramming of human fibroblasts to hepatocyte-like cells was proposed to generate large-scale functional hepatocytes demanded by liver tissue engineering. However, the difficulty in obtaining large quantities of human fibroblasts greatly restricted the extensive implementation of this approach. Meanwhile, human umbilical cord mesenchymal stem cells (HUMSCs) are the preferred cell source for HLCs with the advantages of limited ethical concerns, easy accessibility, and propagation in vitro. However, no direct reprogramming protocol for converting HUMSCs to hepatoblast-like cells (HLCs) has been reported. METHODS: HLCs were successfully generated from HUMSCs by forced expression of FOXA3, HNF1A, and HNF4A (collectively as 3TFs) and c-Myc. In vitro and in vivo functional experiments were conducted to demonstrate the hepatic phenotype, characterization, and function of HUMSC-derived HLCs (HUMSC-iHeps). ChIP-seq and RNA-seq were integrated to reveal the potential molecular mechanisms underlying c-Myc-mediated reprogramming. RESULTS: We showed that c-Myc greatly improved the trans-differentiation efficiency for HLCs from HUMSCs, which remained highly efficient in reprogramming fibroblasts into HLCs, suggesting c-Myc could promote direct reprogramming and its potentially widespread applicability for generating large amounts of HLCs in vitro. Mice transplantation experiments further confirmed the therapeutic potential of HUMSC-iHeps by liver function restoration and survival prolongation. Besides, in vivo safety assessment demonstrated the low risk of the tumorigenic potential of HUMSC-iHeps. We found that c-Myc functioned predominantly at an early phase of reprogramming, and we further unraveled the regulatory network altered by c-Myc. CONCLUSIONS: c-Myc enhanced reprogramming efficiency of HLCs from HUMSCs. A large scale of functional HLCs generated more conveniently from HUMSCs could benefit biomedical studies and applications of liver diseases.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Diferenciación Celular , Hepatocitos , Humanos , Ratones , Cordón Umbilical
9.
Stem Cell Res Ther ; 12(1): 294, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-34016164

RESUMEN

BACKGROUND: Liver fibrosis (LF) is a common pathological process characterized by the activation of hepatic stellate cells (HSCs) and accumulation of extracellular matrix. Severe LF causes cirrhosis and even liver failure, a major cause of morbidity and mortality worldwide. Transplantation of human placental mesenchymal stem cells (hPMSCs) has been considered as an alternative therapy. However, the underlying mechanisms and the appropriate time window for hPMSC transplantation are not well understood. METHODS: We established mouse models of CCl4-injured LF and administered hPMSCs at different stages of LF once a week for 2 weeks. The therapeutic effect of hPMSCs on LF was investigated, according to histopathological and blood biochemical analyses. In vitro, the effect of hPMSCs and the secretomes of hPMSCs on the inhibition of activated HSCs was assessed. RNA sequencing (RNA-seq) analysis, real-time PCR array, and western blot were performed to explore possible signaling pathways involved in treatment of LF with hPMSCs. RESULTS: hPMSC treatment notably alleviates experimental hepatic fibrosis, restores liver function, and inhibits inflammation. Furthermore, the therapeutic effect of hPMSCs against mild-to-moderate LF was significantly greater than against severe LF. In vitro, we observed that the hPMSCs as well as the secretomes of hPMSCs were able to decrease the activation of HSCs. Mechanistic dissection studies showed that hPMSC treatment downregulated the expression of fibrosis-related genes, and this was accompanied by the upregulation of Caveolin-1 (Cav1) (p < 0.001). This suggested that the amelioration of LF occurred partly due to the restoration of Cav1 expression in activated HSCs. Upregulation of Cav1 can inhibit the TGF-ß/Smad signaling pathway, mainly by reducing Smad2 phosphorylation, resulting in the inhibition of activated HSCs, whereas this effect could be abated if Cav1 was silenced in advance by siRNAs. CONCLUSIONS: Our findings suggest that hPMSCs could provide multifaceted therapeutic benefits for the treatment of LF, and the TGF-ß/Cav1 pathway might act as a therapeutic target for hPMSCs in the treatment of LF.


Asunto(s)
Células Estrelladas Hepáticas , Células Madre Mesenquimatosas , Animales , Femenino , Humanos , Hígado/patología , Cirrosis Hepática/patología , Cirrosis Hepática/terapia , Ratones , Placenta , Embarazo , Regulación hacia Arriba
10.
Cell Death Dis ; 11(6): 482, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32587258

RESUMEN

Transcription factors are known to mediate the conversion of somatic cells to induced pluripotent stem cells (iPSCs). Transcription factor TFAP2C plays important roles in the regulation of embryonic development and carcinogenesis; however, the roles of Tfap2c in regulating somatic cell reprogramming are not well understood. Here we demonstrate Tfap2c is induced during the generation of iPSCs from mouse fibroblasts and acts as a facilitator for iPSCs formation. Mechanistically, the c-Myc-dependent apoptosis, which is a roadblock to reprogramming, can be significantly mitigated by Tfap2c overexpression. Meanwhile, Tfap2c can greatly promote mesenchymal-to-epithelial transition (MET) at initiation stage of OSKM-induced reprogramming. Further analysis of gene expression and targets of Tfap2c during reprogramming by RNA-sequencing (RNA-seq) and ChIP-qPCR indicates that TFAP2C can promote epithelial gene expression by binding to their promoters directly. Finally, knockdown of E-cadherin (Cdh1), an important downstream target of TFAP2C and a critical regulator of MET antagonizes Tfap2c-mediated reprogramming. Taken together, we conclude that Tfap2c serves as a strong activator for somatic cell reprogramming through promoting the MET and inhibiting c-Myc-dependent apoptosis.


Asunto(s)
Apoptosis , Reprogramación Celular , Transición Epitelial-Mesenquimal , Factor de Transcripción AP-2/metabolismo , Animales , Apoptosis/genética , Secuencia de Bases , Reprogramación Celular/genética , Transición Epitelial-Mesenquimal/genética , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Regulación hacia Arriba/genética
11.
Stem Cell Res Ther ; 9(1): 350, 2018 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-30558659

RESUMEN

BACKGROUND: Ischemic vascular diseases are the major cause of death worldwide. In recent years, endothelial cell (EC)-based approaches to vascular regeneration are increasingly viable strategies for treating ischemic diseases, but their applications are challenged by the difficulties in their efficient generation and stable maintenance. Here, we show an alternative protocol that facilitates the generation of functional and expandable ETS variant 2 (ETV2)-induced endothelial-like cells (EiECs) from human adipose-derived stem cells (hADSCs), providing a potential source of cells for autologous ECs to treat ischemic vascular diseases. METHODS: hADSCs were obtained from fresh human adipose tissue. Passage 3 hADSCs were transduced with doxycycline (DOX)-inducible ETV2 transcription factor; purified ETV2-hADSCs were induced into endothelial-like cells using a two-stage induction culture system composed of small molecule compounds and cell factors. EiECs were evaluated for their surface markers, proliferation, gene expression, secretory capacity, and effects on vascular regeneration in vivo. RESULTS: We found that short-term ETV2 expression combined with TGF-ß inhibition is sufficient for the generation of kinase insert domain receptor (KDR)+ cells from hADSCs within 10 days. KDR+ cells showed immature endothelial characteristics, and they can gradually mature in a chemically defined induction medium at the second stage of induction. Futher studies showed that KDR+ cells deriving EC-like cells could stably self-renew and expand about 106-fold in 1 month, and they exhibited expected genome-wide molecular features of mature ECs. Functionally, these EC-like cells significantly promoted revascularization in a hind limb ischemic model. CONCLUSIONS: We isolated highly purified hADSCs and effectively converted them into functional and expandable endothelial-like cells. Thus, the study may provide an alternative strategy to obtain functional EC-like cells with potential for biomedical and pharmaceutical applications.


Asunto(s)
Adipocitos/metabolismo , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Endoteliales/metabolismo , Medicina Regenerativa/métodos , Adipocitos/citología , Animales , Diferenciación Celular , Células Endoteliales/citología , Humanos , Masculino , Ratones , Ratones Desnudos
12.
J Cell Mol Med ; 22(12): 6190-6201, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30255547

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common and deadly malignant tumors in the world, especially in China. Follistatin-like protein 5 (FSTL5) is a member of the FSTL family, which is involved in cell proliferation, migration, differentiation, and embryo development. We aimed to investigate the function and underlying mechanism of FSTL5 in HCC. FSTL5 expression was determined by immunohistochemistry staining in a liver cancer tissue microarray (TMA) and the correlation between FSTL5 and the prognosis of HCC patients was analysed. Further proliferation assay, colony formation assay, flow cytometry, and xenograft tumor model were performed to investigate the bioeffects of FSTL5 in HCC in vitro and in vivo. We found that FSTL5 expression was downregulated in HCC tissues and positively correlated with the prognosis of patients with HCC at tumor node metastasis stage I/II. Overexpression of FSTL5 efficiently impaired HCC growth both in vivo and in vitro with an exogenous manner. Mechanistic investigation demonstrated that FSTL5 promoted HCC cell apoptosis in a caspase-dependent manner and regulated Bcl-2 family proteins. These results indicate that FSTL5 may be a potential novel target for HCC treatment, and a biomarker for tumor prognosis.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas Relacionadas con la Folistatina/genética , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Anciano , Animales , Apoptosis/genética , Carcinoma Hepatocelular/patología , Caspasas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Pronóstico
13.
Stem Cells ; 35(9): 2060-2070, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28514506

RESUMEN

Autologous adipose tissue or adipose tissue with additive adipose-derived mesenchymal stem cells (ADSCs) is used in the breast reconstruction of breast cancer patients who undergo mastectomy. ADSCs play an important role in the angiogenesis and adipogenesis, which make it much better than other materials. However, ADSCs may promote residual tumor cells to proliferate or metastasize, and the mechanism is still not fully understood. In this study, we demonstrated that human ADSCs (hADSCs) could facilitate tumor cells growth after co-injection with MCF7 and ZR-75-30 breast cancer cells (BCCs) by promoting angiogenesis, but hADSCs showed limited effect on the growth of MDA-MB-231 BCCs. Intriguingly, compared with ZR-75-30 tumor cells, MCF7 tumor cells were more potentially promoted by hADSCs in the aspects of angiogenesis and proliferation. Consistent with this, cytokine and angiogenesis array analyses showed that after co-injection with hADSCs, the CXCL1 and CXCL8 concentration were significantly increased in MCF7 tumor, but only moderately increased in ZR-75-30 tumor and did not increase in MDA-MB-231 tumor. Furthermore, we found that CXCL1/8 were mainly derived from hADSCs and could increase the migration and tube formation of human umbilical vein endothelial cells (HUVECs) by signaling via their receptors CXCR1 and CXCR2. A CXCR1/2-specific antagonist (SCH527123) attenuated the angiogenesis and tumor growth in vivo. Our findings suggest that CXCL1/8 secreted by hADSCs could promote breast cancer angiogenesis and therefore provide better understanding of safety concerns regarding the clinical application of hADSCs and suggestion in further novel therapeutic options. Stem Cells 2017;35:2060-2070.


Asunto(s)
Tejido Adiposo/patología , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Quimiocina CXCL1/metabolismo , Interleucina-8/metabolismo , Células Madre Mesenquimatosas/metabolismo , Neovascularización Patológica/patología , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Separación Celular , Ciclobutanos/farmacología , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Stem Cell Res Ther ; 7(1): 105, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27495937

RESUMEN

BACKGROUND: Liver disease is a major cause of death worldwide. Orthotropic liver transplantation (OLT) represents the only effective treatment for patients with liver failure, but the increasing demand for organs is unfortunately so great that its application is limited. Hepatocyte transplantation is a promising alternative to OLT for the treatment of some liver-based metabolic disorders or acute liver failure. Unfortunately, the lack of donor livers also makes it difficult to obtain enough viable hepatocytes for hepatocyte-based therapies. Currently, a fundamental solution to this key problem is still lacking. Here we show a novel non-transgenic protocol that facilitates the rapid generation of functional induced hepatocytes (iHeps) from human adipose-derived stem cells (hADSCs), providing a source of available cells for autologous hepatocytes to treat liver disease. METHODS: We used collagenase digestion to isolate hADSCs. The surface marker was detected by flow cytometry. The multipotential differentiation potency was detected by induction into adipocytes, osteocytes, and chondrocytes. Passage 3-7 hADSCs were induced into iHeps using an induction culture system composed of small molecule compounds and cell factors. RESULTS: Primary cultured hADSCs presented a fusiform or polygon appearance that became fibroblast-like after passage 3. More than 95 % of the cells expressed the mesenchymal cell markers CD29, CD44, CD166, CD105, and CD90. hADSCs possessed multipotential differentiation towards adipocytes, osteocytes, and chondrocytes. We rapidly induced hADSCs into iHeps within 10 days in vitro; the cellular morphology changed from fusiform to close-connected cubiform, which was similar to hepatocytes. After induction, most of the iHeps co-expressed albumin and alpha-1 antitrypsin; they also expressed mature hepatocyte special genes and achieved the basic functions of hepatocyte. Moreover, iHep transplantation could improve the liver function of acute liver-injured NPG mice and prolong life. CONCLUSIONS: We isolated highly purified hADSCs and rapidly induced them into functional hepatocyte-like cells within 10 days. These results provide a source of available cells for autologous hepatocytes to treat liver disease.


Asunto(s)
Adipocitos/citología , Hepatocitos/citología , Células Madre/citología , Adipocitos/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Femenino , Hepatocitos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Hepatopatías/metabolismo , Hepatopatías/terapia , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteocitos/citología , Osteocitos/metabolismo , Células Madre/metabolismo
15.
Biosci Rep ; 36(5)2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27377320

RESUMEN

To observe the effect of gene expression and tumorigenicity in hybrid cells of human embryonic stem cells (hESCs) and ovarian cancer cells in vitro and in vivo using a mouse model, and to determine its feasibility in reprogramming tumour cells growth and apoptosis, for a potential exploration of the role of hESCs and tumour cells fusion in the management of ovarian cancer. Stable transgenic hESCs (H1) and ovarian cancer cell line OVCAR-3 were established before fusion, and cell fusion system was established to analyse the related indicators. PTEN expression in HO-H1 cells was higher than those in the parental stem cells and lower than those in parental tumour cells; the growth of OV-H1 (RFP+GFP) hybrid cells with double fluorescence expressions were obviously slower than that of human embryonic stem cells and OVCAR-3 ovarian cancer cells. The apoptosis signal of the OV-H1 hybrid cells was significantly higher than that of the hESCs and OVCAR-3 ovarian cancer cells. In vivo results showed that compared with 7 days, 28 days and 35 days after inoculation of OV-H1 hybrid cells; also, apoptotic cell detection indicated that much stronger apoptotic signal was found in OV-H1 hybrid cells inoculated mouse. The hESCs can inhibit the growth of OVCAR-3 cells in vitro by suppressing p53 and PTEN expression to suppress the growth of tumour that may be achieved by inducing apoptosis of OVCAR-3 cells. The change of epigenetics after fusion of ovarian cancer cells and hESCs may become a novel direction for treatment of ovarian cancer.


Asunto(s)
Epigénesis Genética , Células Madre Embrionarias Humanas/citología , Células Híbridas/patología , Neoplasias Ováricas/patología , Animales , Apoptosis/genética , Fusión Celular , Línea Celular Tumoral , Proliferación Celular/genética , Resistencia a Antineoplásicos , Femenino , Regulación Neoplásica de la Expresión Génica , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Híbridas/metabolismo , Ratones , Neoplasias Ováricas/metabolismo , Fosfohidrolasa PTEN/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Stem Cell Res Ther ; 6: 193, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26438426

RESUMEN

The generation of functional hepatocytes is a major challenge for regenerative medicine and drug discovery. Here we show a method that facilitates generation of induced functional hepatocytes (iHeps) from adipose-derived stem cells (ADSCs) within 9 days. iHeps express hepatocytic gene programs and display functions characteristic of mature hepatocytes, including cytochrome P450 enzyme activity. Upon transplantation into mice with carbon tetrachloride (CCl4)-induced acute fulminant liver failure, iHeps restore the liver function and prolong survival. The work could contribute to the development of alternative strategies to obtain nonhepatic cell-derived mature hepatocytes with potential for biomedical and pharmaceutical applications.


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular , Hepatocitos/fisiología , Grasa Abdominal/citología , Animales , Tetracloruro de Carbono , Células Cultivadas , Femenino , Hepatocitos/trasplante , Fallo Hepático/inducido químicamente , Fallo Hepático/terapia , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Ratas Endogámicas Lew
17.
Biomaterials ; 34(6): 1701-11, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23218839

RESUMEN

Pluripotent stem cells, including embryonic stem (ES) and induced pluripotent stem (iPS) cells, serve as unlimited resources for cell replacement therapy and tissue engineering because such cells are capable of extensive proliferation in vitro and can give rise to lineages that represent any of the three embryonic germ layers. However, in the context of the in vivo behavior of cell transplants, key challenges need to be addressed and essential strategies should be developed before stem cells can be used in clinical practice. In the present study, we modified mouse ES/iPS cells to contain a suicide gene, deltaTK or CodA, under the transcriptional control of the EF1α or Nanog promoter. The suicide gene was introduced via lentivirus transduction without interfering with their self-renewal and pluripotency characteristics. We found that EF1α promoter-controlled deltaTK/CodA expression efficiently eliminated pluripotent stem cells and their derivatives both in vitro and in vivo. When the suicide gene was under the control of the Nanog promoter, tumor-initiating undifferentiated pluripotent stem cells were selectively ablated in vitro after prodrug treatment. These results indicate that modification of pluripotent stem cells with a suicide gene prior to transplantation offers a safe manner by which wayward stem cells, and their progeny, can be controlled in vivo. Our approach will render the clinical application of human pluripotent stem cells increasingly possible.


Asunto(s)
Neoplasias/patología , Células Madre Pluripotentes/citología , Animales , Secuencia de Bases , Muerte Celular , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Biomaterials ; 33(11): 3195-204, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22269649

RESUMEN

The generation of human induced pluripotent stem cells (hiPSCs) opens a prospect for regenerative medicine. However, transplantation of somatic cells derived from hiPSCs still harbor many risks such as cells' incorrect differentiation or over-proliferation, and the worst, tumor formation. Therefore, it's essential to ravel out these obstacles before their clinical application. Herein, we genetically modified hiPSCs and human embryonic stem cells (hESCs) with a truncated herpes simplex virus delta thymidine kinase (deltaTK) gene driven by EF1α or Nanog promoter to selectively ablate wayward pluripotent stem cells. The results showed that insertion of deltaTK gene did not alter their pluripotency and self-renewal capacity but rendered them sensitive to ganciclovir, which induced elimination of deltaTK(+) cells in vitro in a dose and time-dependent manner, most importantly, facilitated both prevention and ablation of tumors in vivo. Furthermore, comparative analysis between transduced hiPSCs and hESCs showed that there was no difference in ganciclovir sensitivity between them. This approach may help to develop safety strategies for clinical application of hiPSCs in regenerative medicine in the future.


Asunto(s)
Transformación Celular Neoplásica/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Genes Transgénicos Suicidas/genética , Mejoramiento Genético/métodos , Timidina Quinasa/genética , Animales , Apoptosis/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones SCID , Proteína Homeótica Nanog
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...