Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-39000343

RESUMO

Mesenchymal stem cells (MSCs) have shown great potential for the treatment of liver injuries, and the therapeutic efficacy greatly depends on their homing to the site of injury. In the present study, we detected significant upregulation of hepatocyte growth factor (HGF) in the serum and liver in mice with acute or chronic liver injury. In vitro study revealed that upregulation of miR-9-5p or miR-221-3p promoted the migration of human MSCs (hMSCs) toward HGF. Moreover, overexpression of miR-9-5p or miR-221-3p promoted hMSC homing to the injured liver and resulted in significantly higher engraftment upon peripheral infusion. hMSCs reduced hepatic necrosis and inflammatory infiltration but showed little effect on extracellular matrix (ECM) deposition. By contrast, hMSCs overexpressing miR-9-5p or miR-221-3p resulted in not only less centrilobular necrosis and venous congestion but also a significant reduction of ECM deposition, leading to obvious improvement of hepatocyte morphology and alleviation of fibrosis around central vein and portal triads. Further studies showed that hMSCs inhibited the activation of hepatic stellate cells (HSCs) but could not decrease the expression of TIMP-1 upon acute injury and the expression of MCP-1 and TIMP-1 upon chronic injury, while hMSCs overexpressing miR-9-5p or miR-221-3p led to further inactivation of HSCs and downregulation of all three fibrogenic and proinflammatory factors TGF-ß, MCP-1, and TIMP-1 upon both acute and chronic injuries. Overexpression of miR-9-5p or miR-221-3p significantly downregulated the expression of α-SMA and Col-1α1 in activated human hepatic stellate cell line LX-2, suggesting that miR-9-5p and miR-221-3p may partially contribute to the alleviation of liver injury by preventing HSC activation and collagen expression, shedding light on improving the therapeutic efficacy of hMSCs via microRNA modification.


Assuntos
Células Estreladas do Fígado , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , MicroRNAs , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Células Estreladas do Fígado/metabolismo , Animais , Camundongos , Transplante de Células-Tronco Mesenquimais/métodos , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/terapia , Doença Hepática Induzida por Substâncias e Drogas/genética , Masculino , Tetracloreto de Carbono/efeitos adversos , Fator de Crescimento de Hepatócito/metabolismo , Fator de Crescimento de Hepatócito/genética , Camundongos Endogâmicos C57BL , Movimento Celular
2.
Int Immunopharmacol ; 134: 112255, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38744176

RESUMO

Inflammatory bowel disease (IBD) is distinguished by persistent immune-mediated inflammation of the gastrointestinal tract. Previous experimental investigations have shown encouraging outcomes for the use of mesenchymal stem cell (MSC)-based therapy in the treatment of IBD. However, as a primary medication for IBD patients, there is limited information regarding the potential interaction between 5-aminosalicylates (5-ASA) and MSCs. In this present study, we employed the dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mouse model to examine the influence of a combination of MSCs and 5-ASA on the development of UC. The mice were subjected to weight measurement, DAI scoring, assessment of calprotectin expression, and collection of colons for histological examination. The findings revealed that both 5-ASA and MSCs have demonstrated efficacy in the treatment of UC. However, it is noteworthy that 5-ASA exhibits a quicker onset of action, while MSCs demonstrate more advantageous and enduring therapeutic effects. Additionally, the combination of 5-ASA and MSC treatment shows a less favorable efficacy compared to the MSCs alone group. Moreover, our study conducted in vitro revealed that 5-ASA could promote MSC migration, but it could also inhibit MSC proliferation, induce apoptosis, overexpress inflammatory factors (IL-2, IL-12P70, and TNF-α), and reduce the expression of PD-L1 and PD-L2. Furthermore, a significant decrease in the viability of MSCs within the colon was observed as a result of 5-ASA induction. These findings collectively indicate that the use of 5-ASA has the potential to interfere with the therapeutic efficacy of MSC transplantation for the treatment of IBD.


Assuntos
Colite Ulcerativa , Sulfato de Dextrana , Modelos Animais de Doenças , Mesalamina , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Colite Ulcerativa/terapia , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Colite Ulcerativa/induzido quimicamente , Mesalamina/farmacologia , Mesalamina/uso terapêutico , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Humanos , Camundongos Endogâmicos C57BL , Colo/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Células Cultivadas , Masculino , Proliferação de Células/efeitos dos fármacos , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico
3.
Stem Cells ; 42(6): 567-579, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38469899

RESUMO

Wnt/ß-catenin signaling plays a crucial role in the migration of mesenchymal stem cells (MSCs). However, our study has revealed an intriguing phenomenon where Dickkopf-1 (DKK1), an inhibitor of Wnt/ß-catenin signaling, promotes MSC migration at certain concentrations ranging from 25 to 100 ng/mL while inhibiting Wnt3a-induced MSC migration at a higher concentration (400 ng/mL). Interestingly, DKK1 consistently inhibited Wnt3a-induced phosphorylation of LRP6 at all concentrations. We further identified cytoskeleton-associated protein 4 (CKAP4), another DKK1 receptor, to be localized on the cell membrane of MSCs. Overexpressing the CRD2 deletion mutant of DKK1 (ΔCRD2), which selectively binds to CKAP4, promoted the accumulation of active ß-catenin (ABC), the phosphorylation of AKT (Ser473) and the migration of MSCs, suggesting that DKK1 may activate Wnt/ß-catenin signaling via the CKAP4/PI3K/AKT cascade. We also investigated the effect of the CKAP4 intracellular domain mutant (CKAP4-P/A) that failed to activate the PI3K/AKT pathway and found that CKAP4-P/A suppressed DKK1 (100 ng/mL)-induced AKT activation, ABC accumulation, and MSC migration. Moreover, CKAP4-P/A significantly weakened the inhibitory effects of DKK1 (400 ng/mL) on Wnt3a-induced MSC migration and Wnt/ß-catenin signaling. Based on these findings, we propose that DKK1 may activate the PI3K/AKT pathway via CKAP4 to balance the inhibitory effect on Wnt/ß-catenin signaling and thus regulate Wnt3a-induced migration of MSCs. Our study reveals a previously unrecognized role of DKK1 in regulating MSC migration, highlighting the importance of CKAP4 and PI3K/AKT pathways in this process.


Assuntos
Movimento Celular , Peptídeos e Proteínas de Sinalização Intercelular , Células-Tronco Mesenquimais , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Via de Sinalização Wnt , Proteína Wnt3A , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Movimento Celular/efeitos dos fármacos , Proteína Wnt3A/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Humanos , Animais , beta Catenina/metabolismo , Fosforilação/efeitos dos fármacos , Camundongos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética
4.
Front Nutr ; 10: 1032048, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37006929

RESUMO

Background: Although the association of zinc (Zn) with cardiovascular disease (CVD) has been studied, no consensus has been reached on this relationship, particularly dietary Zn intake. The purpose of this study was to assess the effect of dietary Zn intake on the risk of CVD and to analyze whether this effect varied according to zinc consumption using representative data from China. Methods: 11,470 adults from the China Health and Nutrition Survey (CHNS) were eventually enrolled. The dietary information was collected by the 3 day 24-h dietary recalls combined with dietary weighting method. CVD was defined as participants with self-reported physician-diagnosed apoplexy and/or myocardial infarction during the follow-up. Cox regression was used to calculate the hazard ratios (HRs) of CVD with 95% confidence intervals. Restricted cubic spline function plus Cox regression was used to visualize the influence trend of dietary Zn intake on new-onset CVD and to test whether this trend is linear. 2-segment Cox regression was established to address the nonlinear trend. Results: 431 participants developed CVD, including 262 strokes and 197 myocardial infarctions. Compared with the lowest quintile (Q1), the adjusted hazard ratios and 95% confidence interval (CI) of CVD in Q2 to Q5 of dietary Zn intake were 0.72 (0.54, 0.97), 0.59 (0.42, 0.81), 0.50 (0.34, 0.72) and 0.44 (0.27, 0.71), respectively. The influence trend of dietary Zn intake on new-onset CVD was nonlinear and L-shaped. When dietary Zn intake <13.66 mg/day, increased dietary Zn intake was significantly associated with decreased risk of developing CVD (HR = 0.87, 95% CI: 0.82-0.92, p-value <0.0001). Conclusion: An L-shaped trend was observed between dietary Zn intake and the risk of developing CVD, indicating that dietary Zn intake should be improved moderately, but not excessively, for the benefit of cardiovascular disease.

5.
Stem Cells ; 41(6): 628-642, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-36951300

RESUMO

Migration of mesenchymal stem cells (MSCs) to the site of injury is crucial in transplantation therapy. Studies have shown that cell migration is regulated by the cellular microenvironment and accompanied by changes in cellular metabolism. However, limited information is available about the relationship between MSC migration and cellular metabolism. Here, we show that basic fibroblast growth factor (bFGF) promotes the migration of MSCs with high levels of glycolysis and high expression of hexokinase 2 (HK2), a rate-limiting enzyme in glycolysis. The enhancement of glycolysis via the activation of HK2 expression promoted the migration of MSCs, whereas the inhibition of glycolysis, but not of oxidative phosphorylation, inhibited the bFGF-induced migration of these cells. Furthermore, bFGF enhanced glycolysis by increasing HK2 expression, which consequently promoted ß-catenin accumulation, and the inhibition of glycolysis inhibited the bFGF-induced accumulation of ß-catenin. When the accumulation of glycolytic intermediates was altered, phosphoenolpyruvate was found to be directly involved in the regulation of ß-catenin expression and activation, suggesting that bFGF regulates ß-catenin signaling through glycolytic intermediates. Moreover, transplantation with HK2-overexpressing MSCs significantly improved the effect of cell therapy on skull injury in rats. In conclusion, we propose a novel glycolysis-dependent ß-catenin signaling regulatory mechanism and provide an experimental and theoretical basis for the clinical application of MSCs.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Células-Tronco Mesenquimais , Animais , Ratos , beta Catenina/metabolismo , Movimento Celular , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glicólise , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt
6.
Stem Cell Rev Rep ; 19(2): 358-367, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36242721

RESUMO

Satellite glial cells (SGCs) that surround sensory neurons in the peripheral nervous system ganglia originate from neural crest cells. Although several studies have focused on SGCs, the origin and characteristics of SGCs are unknown, and their lineage remains unidentified. Traditionally, it has been considered that SGCs regulate the environment around neurons under pathological conditions, and perform functions of supporting, nourishing, and protecting neurons. However, recent studies demonstrated that SGCs may have the characteristics of stem cells. After nerve injury, SGCs up-regulate the expression of stem cell markers and can differentiate into functional sensory neurons. Moreover, SGCs express several markers of Schwann cell precursors and Schwann cells, such as CDH19, MPZ, PLP1, SOX10, ERBB3, and FABP7. Schwann cell precursors have also been proposed as a potential source of neurons in the peripheral nervous system. The similarity in function and markers suggests that SGCs may represent a subgroup of Schwann cell precursors. Herein, we discuss the roles and functions of SGCs, and the lineage relationship between SGCs and Schwann cell precursors. We also describe a new perspective on the roles and functions of SGCs. In the DRG located on the posterior root of spinal nerves, satellite glial cells wrap around each sensory neuron to form an anatomically and functionally distinct unit with the sensory neurons. Following nerve injury, satellite glial cells up-regulate the expression of progenitor markers, and can differentiate into neurons.


Assuntos
Neuroglia , Células Satélites Perineuronais , Células Satélites Perineuronais/metabolismo , Neuroglia/metabolismo , Células de Schwann , Células Receptoras Sensoriais
7.
Front Public Health ; 10: 1115908, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699906

RESUMO

Background: As an essential trace element in the body, selenium is associated with the development of many diseases. The purpose of this study was to explore the association between dietary selenium intake and new-onset stroke risk in Chinese adults. Methods: Adults aged ≥18 years in the China Health and Nutrition Survey (CHNS) from 2004 to 2015 were enrolled. Participants were divided into five groups according to the quintile of dietary selenium intake: Q1 (≤ 29.80 µg/day), Q2 (29.80-38.53 µg/day), Q3 (38.53-47.23 µg/day), Q4 (47.23-60.38 µg/day), Q 5(>60.38 µg/day). Cox proportional-hazards model was used to explore the effect of dietary selenium on new-onset stroke. Restricted cubic spline (RCS) was used to visualize the dose-response relationship between dietary selenium and the risk of morbidity. Results: A total of 11,532 subjects were included, and 271 (2.35%) of them developed stroke during a mean follow-up of 6.78 person-years. Compared with the lowest selenium intake group, the HR and 95%CI of stroke in the participants with selenium intake of Q2, Q3, Q4 and Q5 were: 0.85 (0.59, 1.21), 0.62 (0.42, 0.92), 0.43 (0.28, 0.68), 0.49 (0.30, 0.82), respectively. There was an L-shaped relationship between dietary selenium and stroke (nonlinear P-value = 0.0420). The HR and 95%CI of developing stroke was 0.75 (0.65, 0.87) in participants with selenium intake ≤ 60 µg/day. Conclusions: The L-shaped negative association between dietary selenium and stroke in Chinese adults which indicated that dietary selenium should be improved to a certain level to prevent stroke.


Assuntos
Selênio , Acidente Vascular Cerebral , Adulto , Humanos , Adolescente , Estudos Retrospectivos , Acidente Vascular Cerebral/epidemiologia , Inquéritos Nutricionais , China/epidemiologia
8.
Stem Cell Rev Rep ; 17(3): 999-1013, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33389681

RESUMO

Dorsal root ganglia (DRG) sensory neurons can transmit information about noxious stimulus to cerebral cortex via spinal cord, and play an important role in the pain pathway. Alterations of the pain pathway lead to CIPA (congenital insensitivity to pain with anhidrosis) or chronic pain. Accumulating evidence demonstrates that nerve damage leads to the regeneration of neurons in DRG, which may contribute to pain modulation in feedback. Therefore, exploring the regeneration process of DRG neurons would provide a new understanding to the persistent pathological stimulation and contribute to reshape the somatosensory function. It has been reported that a subpopulation of satellite glial cells (SGCs) express Nestin and p75, and could differentiate into glial cells and neurons, suggesting that SGCs may have differentiation plasticity. Our results in the present study show that DRG-derived SGCs (DRG-SGCs) highly express neural crest cell markers Nestin, Sox2, Sox10, and p75, and differentiate into nociceptive sensory neurons in the presence of histone deacetylase inhibitor VPA, Wnt pathway activator CHIR99021, Notch pathway inhibitor RO4929097, and FGF pathway inhibitor SU5402. The nociceptive sensory neurons express multiple functionally-related genes (SCN9A, SCN10A, SP, Trpv1, and TrpA1) and are able to generate action potentials and voltage-gated Na+ currents. Moreover, we found that these cells exhibited rapid calcium transients in response to capsaicin through binding to the Trpv1 vanilloid receptor, confirming that the DRG-SGC-derived cells are nociceptive sensory neurons. Further, we show that Wnt signaling promotes the differentiation of DRG-SGCs into nociceptive sensory neurons by regulating the expression of specific transcription factor Runx1, while Notch and FGF signaling pathways are involved in the expression of SCN9A. These results demonstrate that DRG-SGCs have stem cell characteristics and can efficiently differentiate into functional nociceptive sensory neurons, shedding light on the clinical treatment of sensory neuron-related diseases.


Assuntos
Nociceptividade , Células Receptoras Sensoriais , Humanos , Canal de Sódio Disparado por Voltagem NAV1.7 , Canal de Sódio Disparado por Voltagem NAV1.8 , Nestina , Neuroglia , Dor
9.
ACS Appl Mater Interfaces ; 12(47): 53021-53028, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33170610

RESUMO

As a well-known electron-withdrawing group, benzo[c][1,2,5]thiadiazole (BT) has been intensively studied and adopted to construct polymer donors with tunable band gaps. However, polymer solar cells (PSCs) with BT-based polymer donors, limited by the weak absorption and inflexible energy level of fullerene derivatives, usually suffer mediocre power conversion efficiencies (PCEs). Here, through subtly tailoring a BT unit with asymmetric fluoro and alkyloxy groups and judiciously pairing a BT-based polymer donor with three narrow band gap non-fullerene acceptors (e.g., IEICO-4F, ITOIC-2F, and IDTCN-O), active layers with complementary absorption spectra, small lowest unoccupied molecular orbital (LUMO) offsets, and preferred morphologies have been achieved. Consequently, PSCs with excellent Jsc values (over 20 mA/cm2) and high PCEs up to 12.33% have been obtained. To the best of our knowledge, the value of 12.33% is among the highest PCEs for BT-based polymers in binary PSCs so far. This work demonstrates that the cooperative effect of energy levels, absorption spectra, and morphologies between the donors and acceptors is crucial for governing the performance of organic photovoltaics.

10.
Chem Asian J ; 15(14): 2198-2202, 2020 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-32484590

RESUMO

Two novel molecules TAP and TAH with pronounced reversible halochromic properties have been synthesized and fully characterized. Their butterfly-like structures have been confirmed through single-crystal X-ray diffraction. Their UV-Vis absorption after protonation dramatically red-shifted with naked-eye-visible color change in a very dilute concentration of 10-5 M. Note that the original color of the solution can be recovered after the neutralization with a base.

11.
Mol Metab ; 34: 16-26, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32180556

RESUMO

OBJECTIVE: Adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) influences hepatic cholesterol transportation. Accumulation of hepatic cholesterol leads to fatty liver disease, which is improved by glucagon-like peptide 1 (GLP-1) in diabetes. Therefore, we analyzed the molecular mechanism in the regulation of hepatic ABCA1 by GLP-1 analogue exendin-4. METHODS: Hepatic ABCA1 expression and transcription were checked by western blotting, real-time polymerase chain reaction (PCR), and luciferase assay in HepG2 cells. Chromatin immunoprecipitation (ChIP) and site-directed mutagenesis were employed to determine transcriptional regulation of the ABCA1 gene. Prolactin regulatory element-binding (PREB)-transgenic mice were generated to access the effect of exendin-4 on improving lipid accumulation caused by a high-fat diet (HFD). RESULTS: Exendin-4 stimulated hepatic ABCA1 expression and transcription via the Ca2+/calmodulin (CaM)-dependent protein kinase kinase/CaM-dependent protein kinase IV (CaMKK/CaMKIV) pathway, whereas GLP-1 receptor antagonist exendin9-39 cancelled this effect. Therefore, exendin-4 decreased hepatic lipid content. ChIP showed that PREB could directly bind to the ABCA1 promoter, which was enhanced by exendin-4. Moreover, PREB stimulated ABCA1 promoter activity, and mutation of PREB-binding site in ABCA1 promoter cancelled exendin-4-enhanced ABCA1 promoter activity. Silencing of PREB attenuated the effect of exendin-4 and induced hepatic cholesterol accumulation. Blockade of CaMKK by STO-609 or siRNA cancelled the upregulation of ABCA1 and PREB induced by exendin-4. In vivo, exendin-4 or overexpression of PREB increased hepatic ABCA1 expression and decreased hepatic lipid accumulation and high plasma cholesterol caused by a HFD. CONCLUSIONS: Our data shows that exendin-4 stimulates hepatic ABCA1 expression and decreases lipid accumulation by the CaMKK/CaMKIV/PREB pathway, suggesting that ABCA1 and PREB might be the therapeutic targets in fatty liver disease.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/metabolismo , Peptídeo 1 Semelhante ao Glucagon/agonistas , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Exenatida/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Hep G2 , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Células Tumorais Cultivadas
12.
J Mater Chem B ; 7(47): 7525-7539, 2019 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-31720683

RESUMO

Transplantation of tissue-engineered neural scaffolds bears great potential for reconstructing neural circuits after spinal cord injury (SCI). In this study, a 3D porous silk fibrous scaffold (3D-SF) with biomimetic interconnected micro- to nanofibrous structure and good biocompatibility is fabricated. Then, a small-molecule combination CFLSSVY (CHIR99021, Forskolin, LDN193189, SB431542, SP600125, VPA, and Y27632) that efficiently reprograms rat dermal fibroblasts into neurons is screened, and these chemically induced neurons (CiNs) are shown to readily communicate on the 3D-SF and form neural scaffolds. After transplantation of these silk-based neural scaffolds into the stumps of transected spinal cords in rats, the damaged tissue is repaired significantly, as indicated by the reduced cavity areas, decreased GFAP expression, and improved axonal regeneration and myelination in the injury site. Moreover, the hindlimb movement and motor-nerve conductivity are greatly improved as indicated by the elevated BBB score, the alternate movement of two hindlimbs during the 45° inclined grid test, and the shortened latency and enhanced amplitude in cMEP detection. Together, these results demonstrate that transplantation of neural scaffolds consisting of 3D-SF and dermal fibroblast-reprogrammed neurons leads to significant nerve regeneration and functional recovery, providing a promising therapeutic strategy for SCI.


Assuntos
Nanofibras/química , Neurônios/transplante , Seda/química , Traumatismos da Medula Espinal/terapia , Alicerces Teciduais/química , Animais , Axônios/fisiologia , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Barreira Hematoencefálica/metabolismo , Transdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Reprogramação Celular , Feminino , Fibroblastos/citologia , Humanos , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Porosidade , Ratos , Ratos Sprague-Dawley , Regeneração/fisiologia
13.
Cell Transplant ; 28(12): 1552-1559, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31512502

RESUMO

In this study, we investigated how human umbilical cord mesenchymal stem cells exerted a neuroprotective effect via antiapoptotic mechanisms in a neonatal hypoxic-ischemic encephalopathy rat model. A total of 78 10-day old (P10) rats were used. After human umbilical cord mesenchymal stem cells were collected from human umbilical cords and amplified in culture, they were administered to rat subjects 1 h after induced hypoxic-ischemic encephalopathy treatment. The short-term (48 h) and long-term (28 day) outcomes were evaluated after human umbilical cord mesenchymal stem cells treatment using neurobehavioral function assessment. Triphenyltetrazolium chloride monohydrate staining was performed at 48 h. Beclin-2 and caspase-3 levels were evaluated with Western blot and real time polymerase chain reaction at 48 h. Human umbilical cord mesenchymal stem cells were collected and administrated to hypoxic-ischemic encephalopathy pups by intracerebroventricular injection. Hypoxic-ischemic encephalopathy typically induced significant delay in development and caused impairment in both cognitive and motor functions in rat subjects. Human umbilical cord mesenchymal stem cells were shown to ameliorate hypoxic-ischemic encephalopathy-induced damage and improve both cognitive and motor functions. Although hypoxic-ischemic encephalopathy induced significant expression of caspase-3 and Beclin-2, human umbilical cord mesenchymal stem cells decreased the expression of both of them. Human umbilical cord mesenchymal stem cells may serve as a potential treatment to ameliorate brain injury in hypoxic-ischemic encephalopathy patients.


Assuntos
Hipóxia-Isquemia Encefálica , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Neuroproteção , Cordão Umbilical/metabolismo , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Mesenquimais/patologia , Ratos , Ratos Sprague-Dawley , Cordão Umbilical/patologia
14.
Exp Cell Res ; 379(1): 65-72, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30898547

RESUMO

Olfactory ensheathing cells (OECs) are ideal candidates for cell-based therapies aimed at repairing spinal cord injury (SCI). Accurate targeting of OECs to the lesion site is critical to reconstructing the impaired nervous system. However, the key factors guiding the homing of transplanted OECs to the damaged area after SCI are still unclear. Here, we demonstrate that lysophosphatidic acid (LPA) can significantly facilitate the homing of OECs after SCI in rats. First, we found that OECs exhibited a robust chemotaxis response to LPA in vitro, with LPAR1 being predominant receptor expressed on OECs. We further found that ß-catenin signaling plays an important role in LPA-induced OEC migration. Moreover, silencing LPAR1 not only abolished the migration of OECs but also prevented ERK1/2 phosphorylation and ß-catenin activation, suggesting that LPAR1 ligation serves to activate the ERK1/2 and ß-catenin pathways in LPA-induced OEC chemotactic migration. Finally, cell transplantation experiments confirmed that endogenous LPA, which was observed to be produced at the lesion site after SCI in rat, is a key chemokine that facilitates OEC migration to the injury center. Collectively, our data provide a further description of the homing effects of LPA and a mechanism by which transplanted OECs migrate to the injured area after SCI in rats.


Assuntos
Lisofosfolipídeos/metabolismo , Bulbo Olfatório/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia , Animais , Movimento Celular/fisiologia , Transplante de Células/métodos , Células Cultivadas , Sistema de Sinalização das MAP Quinases/fisiologia , Bulbo Olfatório/citologia , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de Ácidos Lisofosfatídicos/metabolismo , Transdução de Sinais/fisiologia , Medula Espinal/citologia , Medula Espinal/metabolismo , beta Catenina/metabolismo
15.
Stem Cell Rev Rep ; 15(1): 3-12, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30328004

RESUMO

Mesenchymal stem cells (MSCs) represent a promising source of cell-based therapies for treatment of a wide variety of injuries and diseases. Their tropism and migration to the damaged sites, which are elicited by cytokines secreted from tissues around pathology, are the prerequisite for tissue repair and regeneration. Better understanding of the elicited-migration of MSCs and discovering conditions that elevate their migration ability, will help to increase their homing to pathologies and improve therapeutic efficacy. It is increasingly recognized that microRNAs are important regulators of cell migration. Here we summarize current understanding of the microRNA-regulated migration of MSCs.


Assuntos
Movimento Celular/genética , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , Animais , Junções Célula-Matriz , Exossomos/metabolismo , Humanos , MicroRNAs/genética , Transdução de Sinais
16.
Sci Rep ; 8(1): 1986, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29386586

RESUMO

Thyroid stimulating hormone (TSH) stimulates the secretion of thyroid hormones by binding the TSH receptor (TSHR). TSHR is well-known to be expressed in thyroid tissue, excepting it, TSHR has also been expressed in many other tissues. In this study, we have examined the expression of TSHR in rat pancreatic islets and evaluated the role of TSH in regulating pancreas-specific gene expression. TSHR was confirmed to be expressed in rodent pancreatic islets and its cell line, INS-1 cells. TSH directly affected the glucose uptake in INS cells by up-regulating the expression of GLUT2, and furthermore this process was blocked by SB203580, the specific inhibitor of the p38 MAPK signaling pathway. Similarly, TSH stimulated GLUT2 promoter activity, while both a dominant-negative p38MAPK α isoform (p38MAPK α-DN) and the specific inhibitor for p38MAPK α abolished the stimulatory effect of TSH on GLUT2 promoter activity. Finally, INS-1 cells treated with TSH showed increased protein level of glucokinase and enhanced glucose-stimulated insulin secretion. Together, these results confirm that TSHR is expressed in INS-1 cells and rat pancreatic islets, and suggest that activation of the p38MAPK α might be required for TSH-induced GLUT2 gene transcription in pancreatic ß cells.


Assuntos
Transportador de Glucose Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Tireotropina/farmacologia , Animais , Linhagem Celular , Glucoquinase/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 2/genética , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ratos Wistar , Receptores da Tireotropina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Cell Tissue Res ; 372(1): 99-114, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29322249

RESUMO

The migration of mesenchymal stem cells (MSCs) is critical for their use in cell-based therapies. Accumulating evidence suggests that microRNAs are important regulators of MSC migration. Here, we report that the expression of miR-375 was downregulated in MSCs treated with hepatocyte growth factor (HGF), which strongly stimulates the migration of these cells. Overexpression of miR-375 decreased the transfilter migration and the migration velocity of MSCs triggered by HGF. In our efforts to determine the mechanism by which miR-375 affects MSC migration, we found that miR-375 significantly inhibited the activation of Akt by downregulating its phosphorylation at T308 and S473, but had no effect on the activity of mitogen-activated protein kinases. Further, we showed that 3'phosphoinositide-dependent protein kinase-1 (PDK1), an upstream kinase necessary for full activation of Akt, was negatively regulated by miR-375 at the protein level. Moreover, miR-375 suppressed the phosphorylation of focal adhesion kinase (FAK) and paxillin, two important regulators of focal adhesion (FA) assembly and turnover, and decreased the number of FAs at cell periphery. Taken together, our results demonstrate that miR-375 inhibits HGF-elicited migration of MSCs through downregulating the expression of PDK1 and suppressing the activation of Akt, as well as influencing the tyrosine phosphorylation of FAK and paxillin and FA periphery distribution.


Assuntos
Movimento Celular/efeitos dos fármacos , Regulação para Baixo , Fator de Crescimento de Hepatócito/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/metabolismo , Animais , Sequência de Bases , Regulação para Baixo/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Células HEK293 , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , MicroRNAs/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Paxilina/metabolismo , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
18.
Sci Rep ; 7(1): 10013, 2017 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-28855566

RESUMO

Directed migration of the transplanted mesenchymal stem cells (MSCs) to the lesion sites plays a pivotal role in the efficacy of cell-based therapy. Our previous study demonstrates that MSCs under varying neural differentiation states possess different migratory capacities in response to chemoattractants. However, the underlying mechanism has not been fully addressed. Herein, we show that the assembly and turnover of focal adhesions, the phosphorylation of FAK and paxillin, and the reorganisation of F-actin in MSCs are closely related to their differentiation states in response to SDF-1α. Upon SDF-1α stimulation, FAs turnover more rapidly with the most obvious reduction in the existing time of FAs in MSCs of 24-h preinduction that exhibit the most effective migration towards SDF-1α. Further, we confirm that PI3K/Akt and MAPK pathways participate in the regulation of SDF-1α-induced cell migration and FA assembly, and moreover, that the regulatory effects vary greatly depending on the differentiation states. Collectively, these results demonstrate that FA assembly and turnover, which is accompanied with F-actin reorganisation in response to SDF-1α, correlates closely with the differentiation states of MSCs, which might contribute to the different chemotactic responses of these cells, and thus help develop new strategy to improve the efficacy of MSCs-based therapy.


Assuntos
Quimiocina CXCL12/metabolismo , Adesões Focais/metabolismo , Células-Tronco Mesenquimais/parasitologia , Actinas/metabolismo , Animais , Movimento Celular , Quimiotaxia , Quinase 1 de Adesão Focal/metabolismo , Paxilina/metabolismo , Fosforilação , Multimerização Proteica , Processamento de Proteína Pós-Traducional , Ratos Sprague-Dawley , Transdução de Sinais
19.
Stem Cells Int ; 2017: 5251313, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28630630

RESUMO

Spinal cord injury (SCI) represents one of the most complicated and heterogeneous pathological processes of central nervous system (CNS) impairments, which is still beyond functional regeneration. Transplantation of mesenchymal stem cells (MSCs) has been shown to promote the repair of the injured spinal cord tissues in animal models, and therefore, there is much interest in the clinical use of these cells. However, many questions which are essential to improve the therapy effects remain unanswered. For instance, the functional roles and related molecular regulatory mechanisms of MSCs in vivo are not yet completely determined. It is important for transplanted cells to migrate into the injured tissue, to survive and undergo neural differentiation, or to play neural protection roles by various mechanisms after SCI. In this review, we will focus on some of the recent knowledge about the biological behavior and function of MSCs in SCI. Meanwhile, we highlight the function of biomaterials to direct the behavior of MSCs based on our series of work on silk fibroin biomaterials and attempt to emphasize combinational strategies such as tissue engineering for functional improvement of SCI.

20.
Am J Physiol Cell Physiol ; 313(1): C80-C93, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28424168

RESUMO

Mesenchymal stem cells (MSCs) have the potential to treat various tissue damages, but the very limited number of cells that migrate to the damaged region strongly restricts their therapeutic applications. Full understanding of mechanisms regulating MSC migration will help to improve their migration ability and therapeutic effects. Increasing evidence shows that microRNAs play important roles in the regulation of MSC migration. In the present study, we reported that miR-9-5p was upregulated in hepatocyte growth factor -treated MSCs and in MSCs with high migration ability. Overexpression of miR-9-5p promoted MSC migration, whereas inhibition of endogenous miR-9-5p decreased MSC migration. To elucidate the underlying mechanism, we screened the target genes of miR-9-5p and report for the first time that CK1α and GSK3ß, two inhibitors of ß-catenin signaling pathway, were direct targets of miR-9-5p in MSCs and that overexpression of miR-9-5p upregulated ß-catenin signaling pathway. In line with these data, inhibition of ß-catenin signaling pathway by FH535 decreased the miR-9-5p-promoted migration of MSCs, while activation of ß-catenin signaling pathway by LiCl rescued the impaired migration of MSCs triggered by miR-9-5p inhibitor. Furthermore, the formation and distribution of focal adhesions as well as the reorganization of F-actin were affected by the expression of miR-9-5p. Collectively, these results demonstrate that miR-9-5p promotes MSC migration by upregulating ß-catenin signaling pathway, shedding light on the optimization of MSCs for cell replacement therapy through manipulating the expression level of miR-9-5p.


Assuntos
Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Neurônios/metabolismo , beta Catenina/genética , Actinas/genética , Actinas/metabolismo , Animais , Caseína Quinase I/genética , Caseína Quinase I/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Regulação da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Fator de Crescimento de Hepatócito/farmacologia , Cloreto de Lítio/farmacologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , MicroRNAs/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sulfonamidas/farmacologia , Transfecção , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...