Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Biomed Pharmacother ; 170: 116100, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38159379

RESUMO

The replicative senescence of human amniotic epithelial stem cells (hAECs) is a major concern towards its clinical application. This study found that a 300-kDa hyaluronic acid (HA) could effectively delay the replicative senescence of hAECs, as indicated by the downregulation of cellular senescence markers and alteration of the cell cycle, and substantially improve the differentiation capacities of hAECs. HA was confirmed to regulate the CD44 isoform switch by upregulating the CD44s and downregulating the CD44v, thus exerting an anti-aging effect. We further found that HA induced the upregulation of hyaluronan synthase (HAS) 2, resulting in the activation of epithelial splicing regulatory protein 1 (ESRP1) and alternative splicing of CD44 mRNA, thereby promoting CD44s expression and inhibiting CD44v expression. Knockdown of HAS2 blocked ESRP1 expression and attenuated the anti-aging effects of HA. Hermes-1, a specific blocker of CD44, caused partial loss of the anti-aging effect of HA, upregulated senescence markers, and downregulated stemness markers. Furthermore, CD44s receptor activation was shown to initiate the AKT/mTOR downstream signaling. Conclusively, the study suggested that HA delayed hAEC senescence by regulating CD44 isoform switch to activate the AKT/mTOR signaling pathway, and there is potential for the clinical application of hAECs in combination with HA.


Assuntos
Ácido Hialurônico , Proteínas Proto-Oncogênicas c-akt , Humanos , Ácido Hialurônico/farmacologia , Ácido Hialurônico/metabolismo , Linhagem Celular Tumoral , Isoformas de Proteínas/genética , Fatores de Transcrição , Células-Tronco/metabolismo , Serina-Treonina Quinases TOR , Receptores de Hialuronatos/metabolismo
2.
Biomed Pharmacother ; 170: 116033, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38128181

RESUMO

Although stem cell transplantation is an effective strategy in the treatment of type 1 diabetes mellitus (T1DM), the mechanisms underlying its therapeutic effects remain unclear. We hypothesized that stem cells target gut microbiota and intestinal mucosal immunity to promote therapeutic effects against T1DM. We investigated the effects of human amniotic mesenchymal stem cells (hAMSCs) on intestinal microbiota and mucosal immunity in streptozotocin-induced T1DM mice. hAMSCs promoted significant reductions in blood glucose levels and increased the number of insulin-secreting cells in the T1DM model. Compared with T1DM model mice, 16S rRNA sequencing revealed significant differences in the composition, diversity, and abundance of microbiota in the ileum of hAMSC-treated mice. Bifidobacterium, Prevotella, and Alcaligenes species were among the 15 most abundant differential bacterial species. LC-MS revealed significant changes in ileal metabolites, and among the top 100 differential metabolites identified, we found that a significant increase in taurine was closely associated with hAMSC therapy. Additionally, we detected significant differences between the two groups with respect to the frequency and phenotype of CD4+ T cell subsets in mesenteric lymph nodes, and hAMSCs promoted significant increases in Th2 and Treg cell frequencies and reduced the frequencies of Th1 and Th17 cells. Moreover, correlation analysis revealed pairwise correlations between differential microflora and differential metabolites and immune signatures. hAMSCs thus have positive effects on the microbiota and their metabolites in the ileum and intestinal mucosal immunity in T1DM. Our findings indicate that gut microbiota and intestinal mucosal immunity may play vital roles in the hAMSC-based treatment of T1DM.


Assuntos
Diabetes Mellitus Tipo 1 , Microbioma Gastrointestinal , Humanos , Camundongos , Animais , RNA Ribossômico 16S , Transplante de Células-Tronco
3.
Int J Ophthalmol ; 16(7): 1138-1144, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37465516

RESUMO

AIM: To explore the relationship between ocular and systemic conditions and the impact of ocular complications on the quality of life (QOL) in patients after allogeneic hematopoietic stem cell transplantation (ALLO-HSCT). METHODS: Forty-four patients with severe hematopoietic disease were enrolled after ALLO-HSCT at our center from July 2018 to October 2020. They completed two questionnaires: the Ocular Surface Disease Index (OSDI) and the quality-of-life scale for Chinese patients with visual impairment (SQOL-DV1). Ocular conditions and systemic conditions were also assessed. RESULTS: Eye damage was correlated with total bilirubin (P=0.005), and gamma-glutamyl transferase (GGT) (P=0.021). There was no significant correlation between the overall QOL score and OSDI (P=0.8226) or SQOL-DV1 (P=0.9526) scores. The OSDI and the overall QOL score were not correlated with ocular conditions, including best-corrected visual acuity (BCVA), intraocular pressure, Schirmer tear test II, sodium fluorescein staining, tear film breakup time, and tear meniscus height. SQOL-DV1 was correlated with BCVA (P=0.0007), sodium fluorescein staining (P=0.007), and tear film breakup time (P=0.0146). CONCLUSION: In some patients, early ocular symptoms are not evident after ALLO-HSCT, while ocular surface complications can be observed after a comprehensive ophthalmological examination. Especially for those with elevated total bilirubin or GGT, regular ophthalmic follow-up visits are essential to diagnose and treat ocular graft versus host disease (oGVHD), especially for patients with elevated total bilirubin or GGT.

4.
PeerJ ; 11: e15444, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37309372

RESUMO

Acute inflammation is a beneficial response to the changes caused by pathogens or injuries that can eliminate the source of damage and restore homeostasis in damaged tissues. However, chronic inflammation causes malignant transformation and carcinogenic effects of cells through continuous exposure to pro-inflammatory cytokines and activation of inflammatory signaling pathways. According to the theory of stem cell division, the essential properties of stem cells, including long life span and self-renewal, make them vulnerable to accumulating genetic changes that can lead to cancer. Inflammation drives quiescent stem cells to enter the cell cycle and perform tissue repair functions. However, as cancer likely originates from DNA mutations that accumulate over time via normal stem cell division, inflammation may promote cancer development, even before the stem cells become cancerous. Numerous studies have reported that the mechanisms of inflammation in cancer formation and metastasis are diverse and complex; however, few studies have reviewed how inflammation affects cancer formation from the stem cell source. Based on the stem cell division theory of cancer, this review summarizes how inflammation affects normal stem cells, cancer stem cells, and cancer cells. We conclude that chronic inflammation leads to persistent stem cells activation, which can accumulate DNA damage and ultimately promote cancer. Additionally, inflammation not only facilitates the progression of stem cells into cancer cells, but also plays a positive role in cancer metastasis.


Assuntos
Autorrenovação Celular , Neoplasias , Humanos , Divisão Celular , Inflamação , Células-Tronco Neoplásicas
5.
Photodiagnosis Photodyn Ther ; 42: 103569, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37068646

RESUMO

PURPOSE: To evaluate the radial peripapillary capillary vessel density (RPC-VD) and thickness of the retinal nerve fiber layer (RNFL) in acute leukemia (AL) and the associations of these characteristics with blood laboratory parameters. METHODS: A cross-sectional study was performed at the Ophthalmology Department of the Sun Yat-sen Memorial Hospital from February 2019 to April 2022. Sixty eyes of 30 patients diagnosed with AL and sixty eyes of 30 matched healthy controls were included. Optical coherence tomography angiography (OCTA) in the 4.5-mm Angio Disc scan mode and the Ganglion cell complex scan mode were performed for all participants. Correlation analyses were used to examine the associations of RPC-VD and RNFL with blood laboratory parameters. RESULTS: Patients in the AL group had significantly increased RPC-VD in the whole-image (51.42±0.35 vs. 49.52±0.36) and peripapillary fields (53.90±0.43 vs. 51.17±0.50) compared with people in the control group (all P<0.001), while no difference was found for RPC-VD in the inside optic disk fields in the two groups. The RNFL in the AL group was significantly thicker than that in the control group (131.10±3.89 µm vs. 115.03±2.22 µm, P<0.05). Complete blood count (CBC) parameters, including red blood cells, hemoglobin and hematocrit, had a significant negative correlation with RPC-VD and RNFL (all P <0.05). CONCLUSION: An increased RPC-VD and a thicker RNFL are evidence of fundus changes in patients with early-stage AL, and these metrics may be related to decreases in red blood cells, hemoglobin and hematocrit.


Assuntos
Leucemia Mieloide Aguda , Fotoquimioterapia , Humanos , Angiofluoresceinografia/métodos , Capilares/diagnóstico por imagem , Vasos Retinianos/diagnóstico por imagem , Tomografia de Coerência Óptica/métodos , Estudos Transversais , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes , Doença Aguda
6.
Biotechnol Lett ; 45(3): 411-422, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36680638

RESUMO

OBJECTIVES: CD44 is the major receptor for hyaluronan (HA), but its effect on HA-induced differentiation of human amnion mesenchymal stem cells into chondrocytes is unclear. This study aimed to investigate the effects and mechanisms of CD44 in HA-induced chondrogenesis. METHODS: Immunocytochemistry and toluidine blue staining were used to assess the secretion of type II collagen and aggrecan, respectively. qRT-PCR and western blotting were performed to evaluate the expression of key genes and proteins. RESULTS: The expression of aggrecan and type II collagen was downregulated after using the anti-CD44 antibody (A3D8). The transcriptional levels of chondrocytes­associated genes SRY­box transcription factor 9, aggrecan, and collagen type II alpha 1 chain were also decreased. Thus, CD44 may mediate HA-induced differentiation of hAMSCs into chondrocytes. Further investigation indicated that expression of phosphorylated (p)­Erk1/2 and p­Smad2 decreased following CD44 inhibition. The changes in the expression of p-Erk1/2 and p-Smad2 were consistent after using the ERK1/2 inhibitor (U0126) and agonist (EGF), respectively. After administering the p-Smad2 inhibitor, the expression levels of p-ERK1/2 and p-Smad2 appeared downregulated. The results showed crosstalk between Erk1/2 and Smad2. Moreover, inhibition of p-Erk1/2 and p-Smad2 significantly reduced the accumulation of aggrecan and type II collagen. CONCLUSION: These data indicate that CD44 mediates HA-induced differentiation of hAMSCs into chondrocytes by regulating Erk1/2 and Smad2 signaling.


Assuntos
Condrócitos , Células-Tronco Mesenquimais , Humanos , Condrócitos/metabolismo , Ácido Hialurônico/metabolismo , Agrecanas/metabolismo , Agrecanas/farmacologia , Âmnio , Colágeno Tipo II/genética , Diferenciação Celular , Receptores de Hialuronatos/metabolismo , Condrogênese , Células Cultivadas
7.
BMC Ophthalmol ; 23(1): 28, 2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36690959

RESUMO

OBJECTIVE: To develop and validate a nomogram model for predicting chronic ocular graft-versus-host disease (coGVHD) in patients after allogenic haematopoietic stem cell transplantation (allo-HSCT). METHODS: This study included 61 patients who survived at least 100 days after allo-HSCT. Risk factors for coGVHD were screened using LASSO regression, then the variables selected were subjected to logistic regression. Nomogram was established to further confirm the risk factors for coGVHD. Receiver operating characteristic (ROC) curves were constructed to assess the performance of the predictive model with the training and test sets. Odds ratios and 95% confidence intervals (95% CIs) were calculated by using logistic regression analysis. RESULTS: Among the 61 patients, 38 were diagnosed with coGVHD. We selected five texture features: lymphocytes (LYM) (OR = 2.26), plasma thromboplastin antecedent (PTA) (OR = 1.19), CD3 + CD25 + cells (OR = 1.38), CD3 + HLA-DR + cells (OR = 0.95), and the ocular surface disease index (OSDI) (OR = 1.44). The areas under the ROC curve (AUCs) of the nomogram with the training and test sets were 0.979 (95% CI, 0.895-1.000) and 0.969 (95% CI, 0.846-1.000), respectively.And the Hosmer-Lemeshow test was nonsignificant with the training (p = 0.9949) and test sets (p = 0.9691). CONCLUSION: We constructed a nomogram that can assess the risk of coGVHD in patients after allo-HSCT and help minimize the irreversible loss of vision caused by the disease in high-risk populations.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Humanos , Nomogramas , Transplante Homólogo/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Doença Enxerto-Hospedeiro/etiologia , Fatores de Risco
8.
Scand J Immunol ; 97(3): e13246, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36575914

RESUMO

Stem cell engraftment is currently a promising approach for type 1 diabetes mellitus (T1DM) treatment. In our previous study, engraftment of a combination of human amniotic epithelial cells (hAECs) and hyaluronic acid (HA) showed potent anti-diabetic effect in streptozotocin (STZ)-induced T1DM mice via tail vein injection. Here, we adopted a different route of stem cell delivery, that is via pancreatic subcapsular transplantation. This combined local engraftment of hAECs and HA in STZ-induced T1DM rats showed potent anti-diabetic activity, leading to stronger hypoglycaemia, more intact islet structure and increased number of insulin-positive cells compared with those with hAECs or insulin treatments. Engraftment of hAECs alone increased the proportion of Th1 and T-reg cells and decreased the proportion of Th2 and Th17 cells to protect islet ß cells in STZ-induced T1DM rats, whereas the combined engraftment of hAECs and HA showed more potent regulatory capacity, considerably decreased the level of TNF-α and IL-17 and increased the level of TGF-ß1 compared with those by other treatments. The potent synergistic effect of HA contributed to the recovery of immune balance in the diabetic rat model, thereby suggesting a new strategy for effective treatment of T1DM.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Humanos , Ratos , Camundongos , Animais , Diabetes Mellitus Tipo 1/terapia , Ácido Hialurônico/metabolismo , Ácido Hialurônico/farmacologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Insulina/farmacologia , Células Epiteliais/metabolismo
9.
Eur J Pharmacol ; 935: 175326, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36257381

RESUMO

The regulation of stem cell directional differentiation is a core research topic in regenerative medicine, and modulating the fate of stem cells is a promising strategy for precise intervention through the utilization of naturally small molecule compounds. The present study aimed to explore the potential pro-osteogenic differentiation effect of galangin, a flavonoid derived from Alpinia officinarum, on human amniotic mesenchymal stem cells (hAMSCs) and the underlying molecular mechanism. The results showed that galangin had no cytotoxicity towards hAMSCs when the concentration was less than 50 µM. Treatment with 10 µM galangin significantly increased alkaline phosphatase (ALP) secretion and calcium deposition in hAMSCs. Meanwhile, galangin upregulated the mRNA and protein expression of early osteoblast-specific markers, namely ALP, RUNX2, and OSX, and late osteoblast-specific markers, CoL1α1, OPN, and OCN, in hAMSCs. Furthermore, signaling pathway screening studies showed that galangin enhanced the phosphorylation of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). In addition, molecular docking results suggest there is a promising interaction between galangin and JAK2. Finally, treatment with the JAK2 specific inhibitor AG490 effectively reversed the induction of osteogenic differentiation, upregulation of osteoblast-specific marker expression, and activation of JAK2/STAT3 signaling induced by galangin. These results show that galangin induces the osteogenic differentiation of hAMSCs through the JAK2/STAT3 signaling pathway and could serve as a promising small molecular osteoinducer for application to hAMSCs in regenerative medicine.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo , Simulação de Acoplamento Molecular , Diferenciação Celular , Flavonoides/farmacologia , Flavonoides/metabolismo , Transdução de Sinais
10.
Aging Cell ; 21(9): e13686, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35929187

RESUMO

Stem cell senescence is an important cause of aging. Delaying senescence may present a novel way to combat aging and age-associated diseases. This study provided a mechanistic insight into the protective effect of ganoderic acid D (GA-D) against human amniotic mesenchymal stem cell (hAMSCs) senescence. GA-D, a Ganoderma lucidum-derived triterpenoid, markedly prevented hAMSCs senescence via activating the Ca2+ calmodulin (CaM)/CaM-dependent protein kinase II (CaMKII)/nuclear erythroid 2-related factor 2 (Nrf2) axis, and 14-3-3ε was identified as a target of GA-D. 14-3-3ε-encoding gene (YWHAE) knockdown in hAMSCs reversed the activation of the CaM/CaMKII/Nrf2 signals to attenuate the GA-D anti-aging effect and increase senescence-associated ß-galactosidase (SA-ß-gal), p16 and p21 expression levels, including reactive oxygen species (ROS) production, thereby promoting cell cycle arrest and decreasing differentiation potential. YWHAE overexpression maintained or slightly enhanced the GA-D anti-aging effect. GA-D prevented d-galactose-caused aging in mice by significantly increasing the total antioxidant capacity, as well as superoxide dismutase and glutathione peroxidase activity, and reducing the formation of malondialdehyde, advanced glycation end products, and receptor of advanced glycation end products. Consistent with the protective mechanism of GA-D against hAMSCs senescence, GA-D delayed the senescence of bone-marrow mesenchymal stem cells in this aging model in vivo, reduced SA-ß-gal and ROS production, alleviated cell cycle arrest, and enhanced cell viability and differentiation via regulating 14-3-3ε and CaM/CaMKII/Nrf2 axis. Therefore, GA-D retards hAMSCs senescence by targeting 14-3-3ε to activate the CaM/CaMKII/Nrf2 signaling pathway. Furthermore, the in vivo GA-D anti-aging effect may involve the regulation of stem cell senescence via the same signal axis.


Assuntos
Proteínas 14-3-3 , Células-Tronco Mesenquimais , Transdução de Sinais , Triterpenos , Proteínas 14-3-3/metabolismo , Animais , Antioxidantes/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proliferação de Células , Senescência Celular/genética , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Triterpenos/farmacologia
11.
Phytomedicine ; 99: 154006, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35299029

RESUMO

BACKGROUND: Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by high levels of proinflammatory cytokines and epithelial barrier dysfunction. The root of Ligularia fischeri (Ledeb.) Turcz. is a traditional Chinese medicinal herb with diverse therapeutic properties, which has been successfully used to treat inflammation-related diseases. However, little is known about its effect and mechanism against UC. PURPOSE: To investigate the efficacy and mechanism of L. fischeri root extracts against UC. METHODS: L. fischeri root samples were prepared using the alcohol extraction method and liquid-liquid extraction method. A dextran sodium sulfate-induced UC mouse model and a lipopolysaccharide (LPS)-induced inflammatory cell model were employed in the present study. Cell apoptosis was detected by TUNEL staining, and an enzyme-linked immunosorbent assay was used to quantify the abundance of inflammatory factors in tissues. Hematoxylin and eosin staining and Masson staining were employed to analyze drug toxicity to the liver and kidney. A myeloperoxidase (MPO) assay kit was used to detect neutrophil infiltration in colon tissues. RT-qPCR was then employed to quantify the transcriptional levels of proinflammatory and apoptotic-related genes, while tight junction and apoptosis-related proteins were quantified via western blotting. Gas Chromatography/Mass Spectrometry analysis was then performed to identify the natural compounds in L. fischeri root extracts. RESULTS: The water decoction extract, methanol extract, and especially the chloroform extract (CE) exerted potent therapeutic effects in UC mice. Similar to the positive control group (5-aminosalicylic acid), oral administration of CE (30, 60, and 90 mg/kg/d) elicited distinct therapeutic effects on UC mice in the medium- and high-dose groups. CE decreased disease activity index, histopathological score, and MPO level significantly, and effectively retained the colon length. Furthermore, CE significantly reduced the levels of proinflammatory cytokines, including interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α and enhanced the expression of tight junction proteins, such as zonula occludens (ZO)-1, ZO-2, claudin-1, and occludin, as well as the transcriptional levels of mucins, such as MUC-1 and MUC-2, in UC mice. Notably, CE prevented apoptosis of colonic epithelial cells by up-regulating Bcl-2 and down-regulating Bax. Also, CE inhibited the secretion of pro-inflammatory cytokines and apoptosis in LPS-induced RAW264.7 macrophages via the activation of Bcl-2/Bax signals. CONCLUSIONS: Collectively, L. fischeri root extracts, especially CE, have obvious therapeutic effects against UC. CE reduces inflammation and protects the intestinal epithelial cells and intestinal epithelial barrier via activation of the Bcl-2/Bax signaling pathway, and may be a promising therapeutic agent for UC treatment.

12.
Biomed Pharmacother ; 146: 112605, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35062070

RESUMO

Osteoporosis is a common disease characterized by skeletal fragility and microarchitectural deterioration. However, existing conventional drugs exhibit limited efficacy and can elicit severe adverse effects; moreover, and novel stem cell-based therapies have not exhibited sufficient therapeutic efficacy. Our hypothesis is that an appropriate osteogenic inducer may improve their therapeutic efficacy. In this study, we found that bisdemethoxycurcumin (BDMC) stimulates the differentiation of human amniotic mesenchymal stem cells (hAMSCs) into osteoblasts without inducing cytotoxicity. Here BDMC enhances calcium deposition in hAMSCs, while promoting the expression of early and late markers of osteoblast differentiation, including ALP, runt-related transcription factor 2, osterix, COL1-α1, osteocalcin, and osteopontin at the transcriptional and translational levels. Mechanistically, BDMC was found to activate the JAK2/STAT3 pathway; whereas AG490 (JAK2/STAT3 pathway inhibitor) inhibited BDMC functioning. Subsequently, we found that the combinatorial therapy of BDMC and hAMSC had a positive synergistic effect on osteoporotic mouse model induced by bilateral ovariectomy, including inhibiting bone loss and bone resorption and improving bone micro-architecture. Moreover, BDMC inhibited production of the bone resorption markers C-terminal telopeptide of type I collagen, and tartrate resistant acid phosphatase, while promoting serum levels of bone formation markers OCN, and procollagen I N-terminal propeptide. BDMC also improved liver and kidney function in osteoporotic mouse model. Collectively, BDMC improved osteoporosis by enhancing hAMSC osteogenesis and exhibited a protective effect on liver and kidney function in an osteoporotic mouse model. Hence, BDMC may serve as an effective adjuvant, and combined therapy with hAMSCs is a promising new approach toward osteoporosis treatment.


Assuntos
Diarileptanoides/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/prevenção & controle , Animais , Feminino , Humanos , Camundongos , Ovariectomia/efeitos adversos
13.
Oxid Med Cell Longev ; 2021: 6635460, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34012501

RESUMO

Oxidative stress, a term that describes the imbalance between oxidants and antioxidants, leads to the disruption of redox signals and causes molecular damage. Increased oxidative stress from diverse sources has been implicated in most senescence-related diseases and in aging itself. The Kelch-like ECH-associated protein 1- (Keap1-) nuclear factor-erythroid 2-related factor 2 (Nrf2) system can be used to monitor oxidative stress; Keap1-Nrf2 is closely associated with aging and controls the transcription of multiple antioxidant enzymes. Simultaneously, Keap1-Nrf2 signaling is also modulated by a more complex regulatory network, including phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), protein kinase C, and mitogen-activated protein kinase. This review presents more information on aging-related molecular mechanisms involving Keap1-Nrf2. Furthermore, we highlight several major signals involved in Nrf2 unbinding from Keap1, including cysteine modification of Keap1 and phosphorylation of Nrf2, PI3K/Akt/glycogen synthase kinase 3ß, sequestosome 1, Bach1, and c-Myc. Additionally, we discuss the direct interaction between Keap1-Nrf2 and the mammalian target of rapamycin pathway. In summary, we focus on recent progress in research on the Keap1-Nrf2 system involving oxidative stress and aging, providing an empirical basis for the development of antiaging drugs.


Assuntos
Envelhecimento/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/genética , Animais , Humanos , Ratos
14.
Mol Cell Biochem ; 476(1): 247-259, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32918185

RESUMO

Nuclear factor-E2-related factor 2 (Nrf2) is a key transcription factor known to be involved in maintaining cell redox balance and signal transduction and plays central role in reducing intracellular oxidative stress damage, delaying cell senescence and preventing age-related diseases. However, it has been shown that the level of Nrf2 decreases with age and that the silencing of the Nrf2 gene is associated with the induction of premature senescence. Therefore, a plethora of researchers have focused on elucidating the regulatory mechanism of Nrf2 in the prevention of cell senescence. This complex regulatory mechanism of Nrf2 in the cell senescence process involves coordinated regulation of multiple signaling molecules. After summarizing the function of Nrf2 and its relationship with cell senescence pathway, this review focuses on the recent advances and progress made in elucidating the regulatory mechanism of Nrf2 in the cell senescence process. Additionally, the information collected here may provide insights for further research on Nrf2, in particular, on its regulatory mechanism in the cell senescence process.


Assuntos
Senescência Celular , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/metabolismo , Envelhecimento , Animais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Regulação da Expressão Gênica , Inativação Gênica , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Ligantes , Subunidade p50 de NF-kappa B/metabolismo , Oxirredução , Estresse Oxidativo , Ratos , Proteína Supressora de Tumor p53/metabolismo
15.
Exp Biol Med (Maywood) ; 245(18): 1708-1721, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32878463

RESUMO

This study aimed to elucidate the molecular mechanisms, whereby hyaluronic acid, a main extracellular matrix component of articular cartilage, promotes the chondrogenic differentiation of human amniotic mesenchymal stem cells (hAMSCs). Our previous findings indicated that hyaluronic acid combined with hAMSCs showed a marked therapeutic effect against rat osteoarthritis. In the present study, hyaluronic acid markedly enhanced the expression of chondrocyte-specific markers including Col2α1, Acan, and Sox9 in hAMSCs, with strong synergistic effects on chondrogenic differentiation, in combination with the commonly used inducer, transforming growth factor ß3 (TGF-ß3). Microarray analysis showed that Ras-like protein family member 11B (RASL11B) played a pivotal role in the process of hyaluronic acid-mediated chondrogenesis of hAMSCs. This directional differentiation was significantly inhibited by RASL11B knockdown, but RASL11B overexpression dramatically promoted the expression of Sox9, a master chondrogenesis transcriptional factor, at the levels of transcription and translation. Increased Sox9 expression subsequently resulted in high expression levels of Col2α1 and Acan and the accumulation of cartilage-specific matrix components, such as type 2 collagen and glycosaminoglycans. Moreover, we observed that RASL11B activated the signal molecules such as ERK1/2, and Smad2/3 in the presence of hyaluronic acid during TGF-ß3-induced chondrogenesis of hAMSCs. Taken together, these findings suggest that hyaluronic acid activates the RASL11B gene to potentiate the chondrogenic differentiation of hAMSCs via the activation of Sox9 and ERK/Smad signaling, thus providing a new strategy for cartilage defect repairing by hyaluronic acid-based stem cell therapy.


Assuntos
Âmnio/citologia , Diferenciação Celular/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ácido Hialurônico/farmacologia , Células-Tronco Mesenquimais/citologia , Proteínas Monoméricas de Ligação ao GTP/genética , Fatores de Transcrição SOX9/metabolismo , Diferenciação Celular/genética , Condrogênese/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta3/farmacologia
16.
Oxid Med Cell Longev ; 2020: 8291413, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32774686

RESUMO

Aging is an important risk factor in the occurrence of many chronic diseases. Senescence and exhaustion of adult stem cells are considered as a hallmark of aging in organisms. In this study, a senescent human amniotic mesenchymal stem cell (hAMSC) model subjected to oxidative stress was established in vitro using hydrogen peroxide. We investigated the effects of ganoderic acid D (GA-D), a natural triterpenoid compound produced from Ganoderma lucidum, on hAMSC senescence. GA-D significantly inhibited ß-galactosidase (a senescence-associated marker) formation, in a dose-dependent manner, with doses ranging from 0.1 µM to 10 µM, without inducing cytotoxic side-effects. Furthermore, GA-D markedly inhibited the generation of reactive oxygen species (ROS) and the expression of p21 and p16 proteins, relieved the cell cycle arrest, and enhanced telomerase activity in senescent hAMSCs. Furthermore, GA-D upregulated the expression of phosphorylated protein kinase R- (PKR-) like endoplasmic reticulum kinase (PERK), peroxidase III (PRDX3), and nuclear factor-erythroid 2-related factor (NRF2) and promoted intranuclear transfer of NRF2 in senescent cells. The PERK inhibitor GSK2656157 and/or the NRF2 inhibitor ML385 suppressed the PERK/NRF2 signaling, which was activated by GA-D. They induced a rebound for the generation of ROS and ß-galactosidase-positive cells and attenuated the differentiation capacity. These findings suggest that GA-D retards hAMSC senescence through activation of the PERK/NRF2 signaling pathway and may be a promising candidate for the discovery of antiaging agents.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Triterpenos/uso terapêutico , Envelhecimento , Senescência Celular , Humanos , Estresse Oxidativo , Fatores de Risco , Transdução de Sinais , Triterpenos/farmacologia
17.
Mol Med Rep ; 21(6): 2357-2366, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32236637

RESUMO

CD44 antigen (CD44) is a transmembrane protein found in cell adhesion molecules and is involved in the regulation of various physiological processes in cells. It was hypothesized that CD44 directly affected the chondrogenic differentiation of human amniotic mesenchymal stem cells (hAMSCs). In the present study, the expression of chondrocyte­associated factors was detected in the absence and presence of the antibody blocker anti­CD44 antibody during the chondrogenic differentiation of hAMSCs. Following inhibition of CD44 expression, the transcriptional levels of chondrocyte­associated genes SRY­box transcription factor 9, aggrecan and collagen type II α 1 chain, as well as the production of chondrocyte markers type II collagen and aggrecan were significantly decreased in hAMSCs. Further investigation indicated that there was no significant change in total ERK1/2 expression following inhibition of CD44 expression; however, phosphorylated (p)­ERK1/2 expression was decreased. The expression of p­Smad2/3 was also upregulated following CD44 inhibition. These data indicated that CD44 may affect the differentiation of hAMSCs into chondrocytes by regulating the Smad2/3 and ERK1/2 signaling pathway.


Assuntos
Âmnio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Condrócitos/metabolismo , Receptores de Hialuronatos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Agrecanas/metabolismo , Condrogênese/efeitos dos fármacos , Colágeno Tipo II/metabolismo , Humanos , Receptores de Hialuronatos/genética , Sistema de Sinalização das MAP Quinases , Fosforilação , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
18.
Artigo em Inglês | MEDLINE | ID: mdl-32211385

RESUMO

Osteoarthritis (OA) is one of the most common refractory degenerative articular cartilage diseases. Human amniotic mesenchymal cells (hAMSCs) have emerged as a promising stem cell source for cartilage repair, and hyaluronic acid (HA) has proven to be a versatile regulator for stem cell transplantation. Herein, an effective and straightforward intra-articular injection therapy using a cocktail of hAMSCs and HA was developed to treat knee OA in a rat model. The injured cartilage was remarkably regenerated, yielding results comparable to normal cartilage levels after 56 days of treatment. Both hAMSCs and HA were indispensable organic components in this therapy, in which HA could synergistically enhance the effects of hAMSCs on cartilage repair. The regenerative mechanism was attributed to the fact that the addition of HA comprehensively enhances the activities of hAMSCs, including chondrogenic differentiation, proliferation, colonization, and regenerative modulation. This cocktail paves a new avenue for injection therapy to treat OA, holding the potential to realize rapid clinical translation.

19.
Biomed Pharmacother ; 123: 109807, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31896066

RESUMO

Osteogenic inducers play central roles in effective stem cell-based treatment of bone defects/losses. However, the current routine osteogenic inducer is a cocktail comprising three components that must be improved due to low induction efficiency and side effects. Therefore, there is an urgent need to develop safer and more effective osteoinducers. Herein, we demonstrated the osteogenic effect of Ganoderal A (GD-A), a tetracyclic triterpenoid compound from Ganoderma lucidum. GD-A showed no cytotoxicity toward human amniotic mesenchymal stem cells (hAMSCs) at doses of 0.001-10 µM; furthermore, 0.01 µM GD-A significantly induced the generation of osteoblast-specific markers, such as alkaline phosphatase, and calcium deposition in hAMSCs. At molecular levels, GD-A promoted the expression of multiple osteoblast differentiation markers, such as RUNX2, OSX, OPN, ALP, OCN, and COL1α1. Both Wnt/ß-catenin and BMP/SMAD signaling were shown as active during hAMSC osteodifferentiation. Furthermore, specific blocking of both signals by KYA1797K and SB431542 significantly inhibited alkaline phosphatase secretion and RUNX2 and ALP expression when used alone or in combination. Meanwhile, both signals were also blocked. These findings suggest that GD-A induces hAMSC differentiation into osteoblasts through signaling cross-talk between Wnt/ß-catenin and BMP/SMAD. Taken together, GD-A is a safe, effective, and novel osteoinducer and might be used for stem cell-based therapy for bone defects/losses.


Assuntos
Âmnio/citologia , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Proteínas Smad/metabolismo , Triterpenos/farmacologia , Via de Sinalização Wnt , Diferenciação Celular/genética , Forma Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Modelos Biológicos , Osteogênese/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Triterpenos/química , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética
20.
Exp Cell Res ; 384(2): 111642, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31562862

RESUMO

Our hypothesis is that hyaluronic acid may regulate the differentiation of human amniotic epithelial cells (hAECs) into insulin-producing cells and help the treatment of type 1 diabetes. Herein, a protocol for the stepwise in vitro differentiation of hAECs into functional insulin-producing cells was developed by mimicking the process of pancreas development. Treatment of hAECs with hyaluronic acid enhanced their differentiation of definitive endoderm and pancreatic progenitors. Endodermal markers Sox17 and Foxa2 and pancreatic progenitor markers Pax6, Nkx6.1, and Ngn3 were upregulated an enhanced gene expression in hAECs, but hAECs did not express the ß cell-specific transcription factor Pdx1. Interestingly, hyaluronic acid promoted the expression of major pancreatic development-related genes and proteins after combining with commonly used inducers of stem cells differentiation into insulin-producing cells. This indicated the potent synergistic effects of the combination on hAECs differentiation in vitro. By establishing a multiple injection transplantation strategy via tail vein injections, hAECs transplantation significantly reduced hyperglycemia symptoms, increased the plasma insulin content, and partially repaired the islet structure in type 1 diabetic mice. In particular, the combination of hAECs with hyaluronic acid exhibited a remarkable therapeutic effect compared to both the insulin group and the hAECs alone group. The hAECs' paracrine action and hyaluronic acid co-regulated the local immune response, improved the inflammatory microenvironment in the damaged pancreas of type 1 diabetic mice, and promoted the trans-differentiation of pancreatic α cells into ß cells. These findings suggest that hyaluronic acid is an efficient co-inducer of the differentiation of hAECs into functional insulin-producing cells, and hAECs treatment with hyaluronic acid may be a promising cell-replacement therapeutic approach for the treatment of type 1 diabetes.


Assuntos
Âmnio/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/terapia , Células Epiteliais/efeitos dos fármacos , Ácido Hialurônico/farmacologia , Ativinas/metabolismo , Âmnio/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Células Epiteliais/metabolismo , Humanos , Inflamação/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA