Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 203
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cells ; 42(5): 475-490, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38427800

RESUMO

Cellular senescence significantly affects the proliferative and differentiation capacities of mesenchymal stem cells (MSCs). Identifying key regulators of senescence and exploring potential intervention strategies, including drug-based approaches, are active areas of research. In this context, S-adenosyl-l-methionine (SAM), a critical intermediate in sulfur amino acid metabolism, emerges as a promising candidate for mitigating MSC senescence. In a hydrogen peroxide-induced MSC aging model (100 µM for 2 hours), SAM (50 and 100 µM) was revealed to alleviate the senescence of MSCs, and also attenuated the level of reactive oxygen species and enhanced the adipogenic and osteogenic differentiation in senescent MSCs. In a premature aging mouse model (subcutaneously injected with 150 mg/kg/day d-galactose in the neck and back for 7 weeks), SAM (30 mg/kg/day by gavage for 5 weeks) was shown to delay the overall aging process while increasing the number and thickness of bone trabeculae in the distal femur. Mechanistically, activation of PI3K/AKT signaling and increased phosphorylation of forkhead box O3 (FOXO3a) was proved to be associated with the antisenescence role of SAM. These findings highlight that the PI3K/AKT/FOXO3a axis in MSCs could play a crucial role in MSCs senescence and suggest that SAM may be a potential therapeutic drug for MSCs senescence and related diseases.


Assuntos
Senescência Celular , Proteína Forkhead Box O3 , Células-Tronco Mesenquimais , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , S-Adenosilmetionina , Transdução de Sinais , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Animais , Senescência Celular/efeitos dos fármacos , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Transdução de Sinais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , S-Adenosilmetionina/farmacologia , S-Adenosilmetionina/metabolismo , Camundongos , Diferenciação Celular/efeitos dos fármacos , Masculino , Humanos , Camundongos Endogâmicos C57BL
2.
Nano Lett ; 24(20): 6102-6111, 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38739578

RESUMO

Acute lung injury (ALI) is a severe inflammatory lung disease, with high mortality rates. Early intervention by reactive oxygen species (ROS) scavengers could reduce ROS accumulation, break the inflammation expansion chain in alveolar macrophages (AMs), and avoid irreversible damage to alveolar epithelial and endothelial cells. Here, we reported cell-penetrating R9 peptide-modified triangular DNA origami nanostructures (tDONs-R9) as a novel nebulizable drug that could reach the deep alveolar regions and exhibit an enhanced uptake preference of macrophages. tDONs-R9 suppressed the expression of pro-inflammatory cytokines and drove polarization toward the anti-inflammatory M2 phenotype in macrophages. In the LPS-induced ALI mouse model, treatment with nebulized tDONs-R9 alleviated the overwhelming ROS, pro-inflammatory cytokines, and neutrophil infiltration in the lungs. Our study demonstrates that tDONs-R9 has the potential for ALI treatment, and the programmable DNA origami nanostructures provide a new drug delivery platform for pulmonary disease treatment with high delivery efficiency and biosecurity.


Assuntos
Lesão Pulmonar Aguda , DNA , Nanoestruturas , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Camundongos , DNA/química , Administração por Inalação , Nanoestruturas/química , Espécies Reativas de Oxigênio/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Citocinas/metabolismo , Peptídeos/química , Nebulizadores e Vaporizadores , Peptídeos Penetradores de Células/química , Modelos Animais de Doenças , Lipopolissacarídeos , Sistemas de Liberação de Medicamentos , Células RAW 264.7
3.
Small ; : e2310241, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38441385

RESUMO

The direct use of mesenchymal stem cells (MSCs) as therapeutics for skin injuries is a promising approach, yet it still faces several obstacles, including limited adhesion, retention, and engraftment of stem cells in the wound area, as well as impaired regenerative and healing functions. Here, DNA-based self-assembled composites are reported that can aid the adhesion of MSCs in skin wounds, enhance MSC viability, and accelerate wound closure and re-epithelialization. Rolling-circle amplification (RCA)-derived DNA flowers, equipped with multiple copies of cyclic Arg-Gly-Asp (cRGD) peptides and anti-von Willebrand factor (vWF) aptamers, act as robust scavengers of reactive oxygen species (ROS) and enable synergistic recognition and adhesion to stem cells and damaged vascular endothelial cells. These DNA structure-aided stem cells are retained at localized wound sites, maintain repair function, and promote angiogenesis and growth factor secretion. In both normal and diabetes-prone db/db mice models with excisional skin injuries, facile topical administration of DNA flower-MSCs elicits rapid blood vessel formation and enhances the sealing of the wound edges in a single dose. DNA composite-engineered stem cells warrant further exploration as a new strategy for the treatment of skin and tissue damage.

4.
Cancer Cell Int ; 24(1): 93, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431606

RESUMO

BACKGROUND: Novel therapeutic targets are urgently needed for treating drug-resistant non-small cell lung cancer (NSCLC) and overcoming drug resistance to molecular-targeted therapies. Regulator of G protein signaling 20 (RGS20) is identified as an upregulated factor in many cancers, yet its specific role and the mechanism through which RGS20 functions in NSCLC remain unclear. Our study aimed to identify the role of RGS20 in NSCLC prognosis and delineate associated cellular and molecular pathways. METHODS: Immunohistochemistry and lung cancer tissue microarray were used to verify the expression of RGS20 between NSCLC patients. CCK8 and cell cloning were conducted to determine the proliferation ability of H1299 and Anip973 cells in vitro. Furthermore, Transcriptome sequencing was performed to show enrichment genes and pathways. Immunofluorescence was used to detect the translocation changes of YAP to nucleus. Western blotting demonstrated different expressions of autophagy and the Hippo-PKA signal pathway. In vitro and in vivo experiments verified whether overexpression of RGS20 affect the proliferation and autophagy of NSCLC through regulating the Hippo pathway. RESULTS: The higher RGS20 expression was found to be significantly correlated with a poorer five-year survival rate. Further, RGS20 accelerated cell proliferation by increasing autophagy. Transcriptomic sequencing suggested the involvement of the Hippo signaling pathway in the action of RGS20 in NSCLC. RGS20 activation reduced YAP phosphorylation and facilitated its nuclear translocation. Remarkably, inhibiting Hippo signaling with GA-017 promoted cell proliferation and activated autophagy in RGS20 knock-down cells. However, forskolin, a GPCR activator, increased YAP phosphorylation and reversed the promoting effect of RGS20 in RGS20-overexpressing cells. Lastly, in vivo experiments further confirmed role of RGS20 in aggravating tumorigenicity, as its overexpression increased NSCLC cell proliferation. CONCLUSION: Our findings indicate that RGS20 drives NSCLC cell proliferation by triggering autophagy via the inhibition of PKA-Hippo signaling. These insights support the role of RGS20 as a promising novel molecular marker and a target for future targeted therapies in lung cancer treatment.

5.
BMC Biol ; 20(1): 95, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35501783

RESUMO

BACKGROUND: Beiging of white fat plays an important role in energy metabolism. Beige adipocytes contribute to the regulation of body weight and body temperature through expenditure of chemical energy to produce heat, and they have therefore recently attracted considerable attention as potential targets for therapeutic approaches in metabolic disorders, including obesity. All adipocytes, including beige adipocytes, differentiate from mesenchymal stem cells (MSCs), which may provide an important path for clinical intervention; however, the mechanism of beiging of human adipose cell-derived MSCs is not fully understood. Here, we provide insights on the role of IRISIN, which is known to be secreted by skeletal muscle and promote beiging of white fat. RESULTS: We established an IRISIN-induced mesenchymal stem cell beiging model and found that IRISIN protein interacts with the MSC membrane protein TRPC3. This interaction results in calcium influx and consequential activation of Erk and Akt signaling pathways, which causes phosphorylation of PPARγ. The phosphorylated PPARγ enters the nucleus and binds the UCP1 promoter region. Furthermore, the role of TRPC3 in the beiging of MSCs was largely abolished in Trpc3-/- mice. We additionally demonstrate that the calcium concentration in the brain of mice increases upon IRISIN stimulation, followed by an increase in the content of excitatory amino acids and norepinephrine, while Trpc3-/- mice exhibit the reverse effect. CONCLUSIONS: We found that TRPC3 is a key factor in irisin-induced beiging of MSCs, which may provide a new target pathway in addressing metabolic disorders. Our results additionally suggest that the interaction of irisin with TRPC3 may affect multiple tissues, including the brain.


Assuntos
Células-Tronco Mesenquimais , PPAR gama , Tecido Adiposo Branco/metabolismo , Animais , Cálcio/metabolismo , Metabolismo Energético , Fibronectinas , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Canais de Cátion TRPC
6.
Cytometry A ; 101(8): 675-681, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35524584

RESUMO

Dermal papilla (DP) cells regulate hair follicle epithelial cells and melanocytes by secreting functional factors, playing a key role in hair follicle morphogenesis and hair growth. DP cells can reconstitute new hair follicles and induce hair regeneration, providing a potential therapeutic strategy for treating hair loss. However, current methods for isolating DP cells are either inefficient (physical microdissection) or only applied to genetically labeled mice. We systematically screened for the surface proteins specifically expressed in skin DP using mRNA expression databases. We identified two antibodies against receptors LEPTIN Receptor (LEPR ) and Scavenger Receptor Class A Member 5 (SCARA5) which could specifically label and isolate DP cells by flow cytometry from mice back skin at the growth phase. The sorted LEPR+ cells maintained the DP characteristics after culturing in vitro, expressing DP marker alkaline phosphatase and functional factors including RSPO1/2 and EDN3, the three major DP secretory factors that regulate hair follicle epithelial cells and melanocytes. Furthermore, the low-passage LEPR+ DP cells could reconstitute hair follicles on nude mice using chamber graft assay when combined with epithelial stem cells. The method of isolating functional DP cells we established here lays a solid foundation for developing DP cell-based therapy.


Assuntos
Derme , Receptores para Leptina , Animais , Células Cultivadas , Derme/metabolismo , Cabelo/metabolismo , Folículo Piloso , Camundongos , Camundongos Nus , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Receptores Depuradores Classe A/metabolismo
7.
Cytotherapy ; 24(4): 393-404, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34863626

RESUMO

BACKGROUND AIMS: Given their low immunogenicity, immunoregulatory effects and multiple differentiation capacity, mesenchymal stromal cells (MSCs) have the potential to be used for "off-the-shelf" cell therapy to treat various diseases. However, the allorejection of MSCs indicates that they are not fully immune-privileged. In this study, the authors investigated the immunogenicity of human adipose-derived MSCs (Ad-MSCs) and identified potential immunogenic molecules. METHODS: To evaluate the immunogenicity of human Ad-MSCs in vivo, cells were transplanted into humanized mice (hu-mice), then T-cell infiltration and clearance of human Ad-MSCs were observed by immunofluorescence and bioluminescence imaging. One-way mixed lymphocyte reaction and flow cytometry were performed to evaluate the immunogenicity of human Ad-MSCs in vitro. High-throughput T-cell receptor (TCR) repertoire sequencing and mass spectrometry were applied to identified potential immunogenic molecules. RESULTS: The authors observed that allogeneic Ad-MSCs recruited human T cells and caused faster clearance in hu-mice than non-humanized NOD.Cg-Prkdcscid IL2rgtm1Wjl/SzJ (NSG) mice. The proliferation and activation of T cells were significantly enhanced during in vitro co-culture with human Ad-MSCs. In addition, the level of HLA-II expression on human Ad-MSCs was dramatically increased after co-culture with human peripheral blood mononuclear cells (PBMCs). High-throughput sequencing was applied to analyze the TCR repertoire of the Ad-MSC-recruited T cells to identify dominant TCR CDR3 sequences. Using synthesized TCR CDR3 peptides, the authors identified several potential immunogenic candidates, including alpha-enolase (ENO1). The ENO1 expression level of Ad-MSCs significantly increased after co-culture with PBMCs, whereas ENO1 inhibitor (ENOblock) treatment decreased the expression level of ENO1 and Ad-MSC-induced proliferation of T cells. CONCLUSIONS: The authors' findings improve the understanding of the immunogenicity of human Ad-MSCs and provide a theoretical basis for the safe clinical application of allogeneic MSC therapy.


Assuntos
Biomarcadores Tumorais , Proteínas de Ligação a DNA , Transplante de Células-Tronco Mesenquimais , Fosfopiruvato Hidratase , Proteínas Supressoras de Tumor , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Células Cultivadas , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fosfopiruvato Hidratase/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transplante Homólogo
8.
J Pharmacol Sci ; 148(1): 19-30, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34924126

RESUMO

Hepatic ischemia/reperfusion (I/R) injury contributes to morbidity and mortality during liver resection or transplantation, with limited effective treatments available. Here, we investigated the potential benefits and underlying mechanisms of pterostilbene (Pt), a natural component of blueberries and grapes, in preventing hepatic I/R injury. Male C57BL/6 mice subjected to partial warm hepatic I/R and human hepatocyte cell line L02 cells exposed to anoxia/reoxygenation (A/R) were used as in vivo and in vitro models, respectively. Our findings showed that pretreatment with Pt ameliorated hepatic I/R injury by improving liver histology, decreasing hepatocyte apoptosis, and reducing plasma ALT and AST levels. Likewise, cell apoptosis, mitochondrial membrane dysfunction, and mitochondrial ROS overproduction in L02 cells triggered by the A/R challenge in vitro were reduced due to Pt administration. Mechanistically, Pt treatment efficiently enhanced mitophagy and upregulated PINK1, Parkin, and LC3B expression. Notably, the protective effect of Pt was largely abrogated after cells were transfected with PINK1 siRNA. Moreover, Pt pretreatment promoted hepatocyte proliferation and liver regeneration in the late phase of hepatic I/R. In conclusion, our findings provide evidence that Pt exerts hepatoprotective effects in hepatic I/R injury by upregulating PINK1-mediated mitophagy.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Infarto Hepático/genética , Infarto Hepático/prevenção & controle , Mitofagia/efeitos dos fármacos , Mitofagia/genética , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/prevenção & controle , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Hepatócitos/fisiologia , Humanos , Regeneração Hepática/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
9.
J Cell Mol Med ; 25(21): 10088-10100, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34592781

RESUMO

It was shown that endothelial progenitor cells (EPCs) have bidirectional differentiation potential and thus perform different biological functions. The purpose of this study was to investigate the effects of slight up-regulation of the Kir2.1 channel on EPC transdifferentiation and the potential mechanism on cell function and transformed cell type. First, we found that the slight up-regulation of Kir2.1 expression promoted the expression of the stem cell stemness factors ZFX and NS and inhibited the expression of senescence-associated ß-galactosidase. Further studies showed the slightly increased expression of Kir2.1 could also improve the expression of pericyte molecular markers NG2, PDGFRß and Desmin. Moreover, adenovirus-mediated Kir2.1 overexpression had an enhanced contractile response to norepinephrine of EPCs. These results suggest that the up-regulated expression of the Kir2.1 channel promotes EPC transdifferentiation into a pericyte phenotype. Furthermore, the mechanism of EPC transdifferentiation to mesenchymal cells (pericytes) was found to be closely related to the channel functional activity of Kir2.1 and revealed that this channel could promote EPC EndoMT by activating the Akt/mTOR/Snail signalling pathway. Overall, this study suggested that in the early stage of inflammatory response, regulating the Kir2.1 channel expression affects the biological function of EPCs, thereby determining the maturation and stability of neovascularization.


Assuntos
Diferenciação Celular , Células Progenitoras Endoteliais/metabolismo , Pericitos/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Biomarcadores , Autorrenovação Celular , Células Cultivadas , Senescência Celular , Desmina/metabolismo , Células Progenitoras Endoteliais/citologia , Modelos Biológicos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Pericitos/citologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos , Transdução de Sinais
10.
J Cell Sci ; 129(11): 2182-9, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27252357

RESUMO

Angiogenesis plays crucial roles in various physiological processes including wound healing and tissue repair. It requires a tight interaction between endothelial cells and their surrounding environment. Mesenchymal stem cells (MSCs), one of the non-endothelial cell types present in the perivascular environment, have been shown to secret exosomes to modulate intercellular communications between MSCs and their target cells. In this study, we initially isolated exosomes secreted by human adipose-derived MSCs (adMSC-Exo) and examined their roles in angiogenesis. We found that adMSC-Exo could be taken up by endothelial cells and significantly promote angiogenesis in vitro and in vivo Further study showed that miR-125a was enriched in adMSC-Exo, and repressed the expression of the angiogenic inhibitor delta-like 4 (DLL4) by targeting its 3' untranslated region. Additionally, adMSC-Exo and its exosomal transferred miR-125a could repress DLL4 expression and modulate endothelial cell angiogenesis through promoting formation of endothelial tip cells. In conclusion, our study indicates that adMSC-Exo can transfer miR-125a to endothelial cells and promote angiogenesis by repressing DLL4. adMSC-Exo, as a pro-angiogenic factor, might be a promising candidate for therapeutical tissue repair.


Assuntos
Exossomos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Neovascularização Fisiológica , Proteínas Adaptadoras de Transdução de Sinal , Tecido Adiposo/citologia , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio , Separação Celular , Exossomos/ultraestrutura , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética
11.
Cell Tissue Res ; 372(3): 535-547, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29480458

RESUMO

HUMSCs were isolated, differentiated and characterized in vitro. Both HUMSCs and smooth muscle cells differentiated from HUMSCs were used to fabricate tissue-engineered fascia equivalents. Forty-eight mature female Sprague Dawley rats were randomly assigned to four groups: group A (GynemeshTMPS, n = 12), group B (GynemeshTMPS + HUMSCs; n = 12), group C (GynemeshTMPS + smooth muscle cells differentiated from HUMSCs; n = 12) and group D (GynemeshTMPS + HUMSCs + smooth muscle cells differentiated from HUMSCs; n = 12). The posterior vaginal wall was incised from the introitus and the mesh was then implanted. Three implants of each type were tested at 1, 4, 8 and 12 weeks. Fibrotic remodeling, inflammation, vascularization and tissue regeneration were histologically assessed. The levels of type I and type III collagen were determined. There was no difference in fibrotic remodeling between cell-seeded and unseeded meshes at any time (p > 0.05). At 12 weeks, there did not appear to be fewer inflammatory cells around the filament bundles in the mesh with cells compared with the mesh alone (P > 0.05). Group D showed a trend toward better vascularization at 12 weeks compared with group A (P < 0.05). Twelve weeks after implantation, a thin layer of new tissue growth covered the unseeded scaffold and a thicker layer covered the cell-seeded scaffold (P < 0.05). No significant difference in the ratio of collagen type I/III could be detected among the different groups after 12 weeks (P > 0.05). HUMSCs with differentiated smooth muscle cells might have a potential role in fascia tissue engineering to repair POP in the future.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Diafragma da Pelve/cirurgia , Procedimentos de Cirurgia Plástica/métodos , Cordão Umbilical/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Fáscia/fisiologia , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Multipotentes/citologia , Miócitos de Músculo Liso/citologia , Fenótipo , Ratos Sprague-Dawley , Regeneração , Engenharia Tecidual
12.
Stem Cells ; 35(9): 2060-2070, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28514506

RESUMO

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.


Assuntos
Tecido Adiposo/patologia , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Quimiocina CXCL1/metabolismo , Interleucina-8/metabolismo , Células-Tronco Mesenquimais/metabolismo , Neovascularização Patológica/patologia , Animais , Benzamidas/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Ciclobutanos/farmacologia , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Nus , Neovascularização Fisiológica/efeitos dos fármacos , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Cell Biochem ; 435(1-2): 59-66, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28523512

RESUMO

A key feature of cancer cachexia is the loss of adipose tissue, mainly due to increased lipolysis and an impairment of adipogenesis. Recent findings have shown that cancer exosomes promoted lipolysis in adipose tissue. However, effects of cancer exosomes on adipogenesis were not reported. In this study, we found that lung cancer exosomes could be internalized by human adipose tissue-derived mesenchymal stem cells (hAD-MSCs) and significantly inhibited hAD-MSC adipogenesis as demonstrated by Oil Red O staining and decreased expression of adipogenic-specific genes. Specifically, TGFß signaling pathway was demonstrated to be involved in the inhibitive effects of lung cancer exosomes on hAD-MSC adipogenesis. Additionally, TGFß was detected in A549 exosomes. Herein, this study reports that the effect of lung cancer cell exosomes on hAD-MSC adipogenic differentiation was mediated by TGFß signaling pathway and suggests the involvement of cancer exosomes in weight loss of cancer cachexia.


Assuntos
Adipogenia , Exossomos/metabolismo , Neoplasias Pulmonares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Células A549 , Exossomos/patologia , Humanos , Neoplasias Pulmonares/patologia , Células-Tronco Mesenquimais/patologia , Proteínas de Neoplasias
15.
J Cell Biochem ; 115(10): 1683-91, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24802236

RESUMO

MiRNAs have been identified in various plants and animals where they function in post-transcriptional regulation. Although studies revealed that dexamethasone play a pivotal role in the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs), the identification of specific miRNAs and their regulatory roles in this process remain poorly defined. In this study, microarrays were used to analyze the miRNA expression profile of dexamethasone-induced hBMSCs derived from three donors, and RT-PCRs were used to confirm the microarray results. Nine upregulated miRNAs and seven downregulated miRNAs were identified. The putative target genes of these miRNAs were predicted using bioinformatics analysis. Subsequently, we focused our attention on the functional analysis of an upregulated miRNA, miR-23a. Overexpression of miR-23a inhibited osteogenic differentiation of hBMSCs at the cellular, mRNA, and protein levels. The results of our study provide an experimental basis for further research on miRNAs functions during osteogenic differentiation of dexamethasone-induced hBMSCs.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Osteogênese/genética , Adulto , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , MicroRNAs/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro , Regulação para Cima
16.
Biochem Biophys Res Commun ; 447(3): 383-7, 2014 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-24727452

RESUMO

Mesenchymal stem cells (MSCs) are non-hematopoietic cells with multi-lineage potential, which makes them attractive targets for regenerative medicine applications. Efficient gene transfer into MSCs is essential for basic research in developmental biology and for therapeutic applications involving gene-modification in regenerative medicine. Adenovirus vectors (Advs) can efficiently and transiently introduce an exogenous gene into many cell types via their primary receptors, the coxsackievirus and adenovirus receptors (CARs), but not into MSCs, which lack CAR expression. To overcome this problem, an Adv coated with cationic polymer polyethyleneimine (PEI) was developed. In this study, we demonstrated that PEI coating with an optimal ratio can enhance adenoviral transduction of MSCs without cytotoxicity. We also investigated the physicochemical properties and internalization mechanisms of the PEI-coated Adv. These results could help to evaluate the potentiality of the PEI-coated Adv as a prototype vector for efficient and safe transduction into MSCs.


Assuntos
Adenoviridae/química , Vetores Genéticos/química , Células-Tronco Mesenquimais , Polietilenoimina/química , Transdução Genética/métodos , Adenoviridae/fisiologia , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/fisiologia , Endocitose , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Vírion/química , Vírion/fisiologia , Internalização do Vírus
17.
BMC Cancer ; 14: 616, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25159494

RESUMO

BACKGROUND: The aberrant expression of microRNAs has been demonstrated to play a crucial role in the initiation and progression of hepatocarcinoma. miR-1246 expression in High invasive ability cell line than significantly higher than that in low invasive ability cell line. METHODS: Transwell chambers (8-uM pore size; Costar) were used in the in vitro migration and invison anssay. Dual luciferase reporter gene construct and Dual luciferase reporter assay to identify the target of miR-1246. CADM1 expression was evaluated by immunohistochemistric staining. The clinical manifestations, treatments and survival were collected for statistical analysis. RESULTS: Inhibition of miR-1246 effectively reduced migration and invasion of hepatocellular carcinoma cell lines. Bioinformatics and luciferase reporter assay revealed that miR-1246 specifically targeted the 3'-UTR of Cell adhesion molecule 1 and regulated its expression. Down-regulation of CADM1 enhanced migration and invasion of HCC cell lines. Furthermore, in tumor tissues obtained from liver cancer patients, the expression of miR-1246 was negatively correlated with CADM1 and the high expression of miR-1246 combined with low expression of CADM1 might serve as a risk factor for stage1 liver cancer patients. CONCLUSIONS: Our study showed that miR-1246, by down-regulation CADM1, enhances migration and invasion in HCC cells.


Assuntos
Carcinoma Hepatocelular/patologia , Moléculas de Adesão Celular/metabolismo , Imunoglobulinas/metabolismo , Neoplasias Hepáticas/patologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidade , Molécula 1 de Adesão Celular , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Imunoglobulinas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Fatores de Risco , Análise de Sobrevida
18.
Int J Toxicol ; 33(5): 403-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25136035

RESUMO

The efficacy of stem cell transplantation for promoting recovery of patients with neurological diseases, such as stroke, has been reported in several studies. However, the safety of the intracerebral transplantation of human mesenchymal stem cells (hMSCs) remains unclear. The aim of the study was to evaluate the safety of hMSCs transplanted in cerebrum of Macaca fascicularis and to provide evidence for clinical application. A total of 24 M fascicularis were assigned to 3 groups randomly: low dose (3.0 × 10(5) cells/kg), high dose (2.5 × 10(6) cells/kg), and the control (normal saline [NS]). Human mesenchymal stem cells or NS were injected into each monkey for 2 times, with an interval of 3 weeks. The injection point was located outside of the right putamen, according to a stereotactic map and preoperative magnetic resonance imaging of the monkeys. Animal health, behavior, biophysical and biochemical parameters, and brain neurological function were routinely monitored over a 6-month period posttransplantation, and the histopathologic examinations were also performed. The results showed that local pathologic damage including local tissue necrosis and inflammation was induced after the injection. The damage of low-dose and high-dose groups was greater than that of the control group, yet over time, the damage could be repaired gradually. No major hMSCs-associated changes were induced from other indicators, and the transplantation of hMSCs in monkeys did not affect total immunoglobulin (Ig) M, total IgG, CD3, CD4, or CD8 values. We therefore conclude that transplantation of hMSCs to the cerebrum represents a safe alternative for clinical application of neurological disorders.


Assuntos
Encéfalo/citologia , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Animais , Temperatura Corporal , Peso Corporal , Líquido Cefalorraquidiano/citologia , Ingestão de Alimentos , Feminino , Humanos , Imunidade , Inflamação/etiologia , Inflamação/patologia , Macaca fascicularis , Masculino , Necrose/etiologia , Necrose/patologia , Exame Neurológico , Tamanho do Órgão
19.
Aging Dis ; 15(2): 787-803, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38447216

RESUMO

Cellular senescence is an irreversible and multifaceted process inducing tissue dysfunction and organismal aging, and thus the clearance of senescent cells can prevent or delay the onset of aging-related pathologies. Herein, we developed an augmented photothermal therapy strategy integrated with an antibody against ß2-microglobulin (aB2MG) and an immune adjuvant imiquimod (R837) to effectively accelerate senescent cell apoptosis and clearance under a near-infrared light. With this strategy, the designed CroR@aB2MG enables the targeting of senescent cells and the application of photothermal therapy concomitantly, the initiation of immune clearance subsequently, and finally the realization of protective effects against senescence. Our results showed that the photo-induced heating effect caused senescent cells to quickly undergo apoptosis and the synchronous immune response accelerated the clearance of senescent cells in vitro and in vivo. Therefore, this photoactivated speedy clearing strategy may provide an efficient way for the treatment of senescence-related diseases by eliminating senescent cells with biomaterials.


Assuntos
Anticorpos , Terapia Fototérmica , Senescência Celular , Imunidade
20.
Aging Dis ; 2024 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-38913037

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease that affects the living quality of patients, especially the elderly population. RA-related morbidity and mortality increase significantly with age, while current clinical drugs for RA are far from satisfactory and may have serious side effects. Therefore, the development of new drugs with higher biosafety and efficacy is demanding. Black phosphorus nanosheets (BPNSs) have been widely studied because of their excellent biocompatibility. Here, we focus on the inherent bioactivity of BPNSs, report the potential of BPNSs as a therapeutic drug for RA and elucidate the underlying therapeutic mechanism. We find that BPNSs inhibit autophagy at an early stage via the AMPK-mTOR pathway, switch the energy metabolic pathway to oxidative phosphorylation, increase intracellular ATP levels, suppress apoptosis, reduce inflammation and oxidative stress, and down-regulate senescence-associated secretory phenotype (SASP)-related genes in rheumatoid arthritis synovial fibroblasts (RA-SFs). Further, BPNSs induce the apoptosis of macrophages and promote their transition from the M1 to the M2 phenotype by regulating related cytokines. Significantly, the administration of BPNSs can alleviate key pathological features of RA in mice, revealing great therapeutic potential. This study provides a novel option for treating RA, with BPNSs emerging as a promising therapeutic candidate.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA