Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Transplant ; 33: 9636897231221882, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38205679

RESUMO

The current study aimed to determine the safety profile of intra-articular-injected allogeneic adipose-derived mesenchymal stem cells (ADSCs) GXCPC1 in subjects with knee osteoarthritis (OA) and its preliminary efficacy outcome. The 3 + 3 phase I study was designed with two dose-escalation cohorts: low dose (6.7 × 106 GXCPC1, N = 5) and high dose (4 × 107 GXCPC1, N = 6). The primary endpoint was safety, which was evaluated by recording adverse events throughout the trial; the secondary endpoints included total, pain, stiffness, and function subscales of the Western Ontario and McMaster Universities Arthritis Index (WOMAC), Visual Analogue Scale (VAS) for pain, and 12-Item Short Form (SF-12) health survey questionnaire. The GXCPC1 treatment was found to be safe after 1 year of follow-up with no treatment-related severe adverse events observed. When compared to baseline, subjects in both the low- and high-dose cohorts demonstrated improving trends in pain and knee function after receiving GXCPC1 treatment. Generally, the net change in pain (95% confidence interval (CI) = -7.773 to -2.561t at 12 weeks compared to baseline) and knee function (95% CI = -24.297 to -10.036t at 12 weeks compared to baseline) was better in subjects receiving high-dose GXCPC1. Although this study included a limited number of subjects without a placebo arm, it showed that the intra-articular injection of ADSCs was safe and well-tolerated in subjects with therapeutic alternatives to treat knee OA. However, a larger scale study with an appropriate control would be necessary for clinical efficacy in the following study.


Assuntos
Células-Tronco Mesenquimais , Osteoartrite do Joelho , Humanos , Injeções Intra-Articulares , Osteoartrite do Joelho/terapia , Dor , Projetos Piloto
2.
Int J Mol Sci ; 24(8)2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37108239

RESUMO

Osteoarthritis (OA) is a degenerative disease that causes pain, cartilage deformation, and joint inflammation. Mesenchymal stem cells (MSCs) are potential therapeutic agents for OA treatment. However, the 2D culture of MSCs could potentially affect their characteristics and functionality. In this study, calcium-alginate (Ca-Ag) scaffolds were prepared for human adipose-derived stem cell (hADSC) proliferation with a homemade functionally closed process bioreactor system; the feasibility of cultured hADSC spheres in heterologous stem cell therapy for OA treatment was then evaluated. hADSC spheres were collected from Ca-Ag scaffolds by removing calcium ions via ethylenediaminetetraacetic acid (EDTA) chelation. In this study, 2D-cultured individual hADSCs or hADSC spheres were evaluated for treatment efficacy in a monosodium iodoacetate (MIA)-induced OA rat model. The results of gait analysis and histological sectioning showed that hADSC spheres were more effective at relieving arthritis degeneration. The results of serological and blood element analyses of hADSC-treated rats indicated that the hADSC spheres were a safe treatment in vivo. This study demonstrates that hADSC spheres are a promising treatment for OA and can be applied to other stem cell therapies or regenerative medical treatments.


Assuntos
Células-Tronco Mesenquimais , Osteoartrite , Ratos , Humanos , Animais , Cálcio/efeitos adversos , Alginatos/efeitos adversos , Osteoartrite/induzido quimicamente , Osteoartrite/terapia , Osteoartrite/patologia , Adipócitos/patologia , Modelos Animais de Doenças
3.
Tissue Eng Regen Med ; 19(6): 1295-1310, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36346531

RESUMO

BACKGROUND: Extracellular vesicles (EVs) are derived from internal cellular compartments, and have potential as a diagnostic and therapeutic tool in degenerative disease associated with aging. Mesenchymal stem cells (MSCs) have become a promising tool for functional EVs production. This study investigated the efficacy of EVs and its effect on differentiation capacity. METHODS: The characteristics of MSCs were evaluated by flow cytometry and stem cell differentiation analysis, and a production mode of functional EVs was scaled from MSCs. The concentration and size of EVs were quantitated by Nanoparticle Tracking Analysis (NTA). Western blot analysis was used to assess the protein expression of exosome-specific markers. The effects of MSC-derived EVs were assessed by chondrogenic and adipogenic differentiation analyses and histological observation. RESULTS: The range of the particle size of adipose-derived stem cells (ADSCs)- and Wharton's jelly -MSCs-derived EVs were from 130 to 150 nm as measured by NTA, which showed positive expression of exosomal markers. The chondrogenic induction ability was weakened in the absence of EVs in vitro. Interestingly, after EV administration, type II collagen, a major component in the cartilage extracellular matrix, was upregulated compared to the EV-free condition. Moreover, EVs decreased the lipid accumulation rate during adipogenic induction. CONCLUSION: The results indicated that the production model could facilitate production of effective EVs and further demonstrated the role of MSC-derived EVs in cell differentiation. MSC-derived EVs could be successfully used in cell-free therapy to guide chondrogenic differentiation of ADSC for future clinical applications in cartilage regeneration.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Adipogenia , Condrócitos , Células Cultivadas , Vesículas Extracelulares/metabolismo , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular
4.
Cell Transplant ; 31: 9636897221106995, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36002988

RESUMO

The global population of individuals afflicted with diabetes mellitus has been increasing year by year, and this disease poses a serious threat to human health as well as the economies worldwide. Pancreatic or islet transplantations provide one of the most effective and long-term therapies available to treat diabetes, but the scarcity and quality of pancreatic islets limit their use in treatments. Here, we report the development of a one-step, monolayer culture, and chemical-based protocol that efficiently mediates the differentiation of human adipose-derived stem cells (hADSCs) into insulin-producing cells (IPCs). Our data indicate that hADSCs in monolayer culture that are allowed to differentiate into IPCs are superior to those in suspension cultures with respect to insulin secretion capacity (213-fold increase), cell viability (93.5 ± 3.27% vs. 41.67 ± 13.17%), and response to glucose stimulation. Moreover, the expression of genes associated with pancreatic lineage specification, such as PDX1, ISL1, and INS (encoding insulin), were expressed at significantly higher levels during our differentiation protocol (6-fold for PDX1 and ISL1, 11.5-fold for INS). Importantly, in vivo studies demonstrated that transplantation with IPCs significantly mitigated hyperglycemia in streptozotocin-induced diabetic rats. Our results indicate that this one-step, rapid protocol increases the efficiency of IPC generation and that the chemical-based approach for IPC induction may reduce safety concerns associated with the use of IPCs for clinical applications, thereby providing a safe and effective cell-based treatment for diabetes.


Assuntos
Diabetes Mellitus Experimental , Hiperglicemia , Células Secretoras de Insulina , Animais , Diferenciação Celular/fisiologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Humanos , Hiperglicemia/terapia , Insulina/metabolismo , Ratos , Células-Tronco , Estreptozocina
5.
Cells ; 11(7)2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35406658

RESUMO

Oligodendrocytes are glial cells located in the central nervous system (CNS) that play essential roles in the transmission of nerve signals and in the neuroprotection of myelinated neurons. The dysfunction or loss of oligodendrocytes leads to demyelinating diseases such as multiple sclerosis (MS). To treat demyelinating diseases, the development of a therapy that promotes remyelination is required. In the present study, we established an in vitro method to convert human fibroblasts into induced oligodendrocyte-like cells (iOLCs) in 3 days. The induced cells displayed morphologies and molecular signatures similar to oligodendrocytes after treatment with valproic acid and exposure to the small molecules Y27632, SU9516, and forskolin (FSK). To pursue the development of a cell-free remyelination therapy in vivo, we used a cuprizone-induced demyelinated mouse model. The small molecules (Y27632, SU9516, and FSK) were directly injected into the demyelinated corpus callosum of the mouse brain. This combination of small molecules rescued the demyelination phenotype within two weeks as observed by light and electron microscopy. These results provide a foundation for exploring the development of a treatment for demyelinating diseases via regenerative medicine.


Assuntos
Cuprizona , Doenças Desmielinizantes , Animais , Corpo Caloso , Cuprizona/efeitos adversos , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/genética , Camundongos , Camundongos Endogâmicos C57BL , Oligodendroglia/fisiologia
6.
J Tissue Eng Regen Med ; 16(1): 3-13, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34644444

RESUMO

Current therapy does not provide significant benefits for patients with chronic stroke. Pre-clinical studies suggested that autologous adipose-derived stem cells have benefits for the treatment of chronic stroke. This Phase I open-label study was conducted to demonstrate the safety and efficacy of autologous adipose-derived stem cells (GXNPC1) in chronic stroke. Three patients with chronic stroke were treated with stereotactic implantation of autologous adipose-derived stem cells (1 × 108 cells). The primary endpoints of safety evaluation included adverse events, over a 6 months post-implantation period. The secondary endpoints included improvements in neurological functions. Evolutional change of brain parenchyma was also followed with magnetic resonance imaging (MRI). All three participants improved significantly at 6 months follow-up. The extent of improvement from pre-treatment was: National Institutes of Health Stroke Scale improved 5-15 points, Barthel Index: 25-50 points, Berg balance scale 0-21 points and Fugl-Meyer modified sensation 3-28 points. All three patients had signal change along the implantation tract on MRI one month after surgery. There is no related safety issue through 6 months observation. Clinical measures of neurological symptoms of these patients with chronic stroke improved at 6 months without adverse effects after implantation of autologous adipose-derived stem cells (GXNPC1), which might be correlated with post-implantation changes on brain MRI. Clinical Trial Registration-URL: https://clinicaltrials.gov/ct2/show/NCT02813512?term=ADSC&cond=Stroke&cntry=TW&draw=2&rank=1 Unique identifier: NCT02813512.


Assuntos
AVC Isquêmico , Transplante de Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Tecido Adiposo , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Acidente Vascular Cerebral/terapia , Transplante Autólogo , Resultado do Tratamento
7.
J Mol Med (Berl) ; 99(12): 1815-1827, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34633469

RESUMO

Cognitive impairment is a serious side effect of post-myocardial infarction (MI) course. We have recently demonstrated that human adipose-derived stem cells (hADSCs) ameliorated myocardial injury after MI by attenuating reactive oxygen species (ROS) levels. Here, we studied whether the beneficial effects of intramyocardial hADSC transplantation can extend to the brain and how they may attenuate cognitive dysfunction via modulating ROS after MI. After coronary ligation, male Wistar rats were randomized via an intramyocardial route to receive either vehicle, hADSC transplantation (1 × 106 cells), or the combination of hADSCs and 3-Morpholinosydnonimine (SIN-1, a peroxynitrite donor). Whether hADSCs migrated into the hippocampus was assessed by using human-specific primers in qPCR reactions. Passive avoidance test was used to assess cognitive performance. Postinfarction was associated with increased oxidative stress in the myocardium, circulation, and hippocampus. This was coupled with decreased numbers of dendritic spines as well as a significant downregulation of synaptic plasticity consisting of synaptophysin and PSD95. Step-through latency during passive avoidance test was impaired in vehicle-treated rats after MI. Intramyocardial hADSC injection exerted therapeutic benefits in improving cardiac function and cognitive impairment. None of hADSCs was detected in rat's hippocampus at the 3rd day after intramyocardial injection. The beneficial effects of hADSCs on MI-induced histological and cognitive changes were abolished after adding SIN-1. MI-induced ROS attacked the hippocampus to induce neurodegeneration, resulting in cognitive deficit. The remotely intramyocardial administration of hADSCs has the capacity of improved synaptic neuroplasticity in the hippocampus mediated by ROS, not the cell engraftment, after MI. KEY MESSAGES: Human adipose-derived stem cells (hADSCs) ameliorated injury after myocardial infarction by attenuating reactive oxygen species (ROS) levels. Intramyocardial administration of hADSCs remotely exerted therapeutic benefits in improving cognitive impairment after myocardial infarction. The improved synaptic neuroplasticity in the hippocampus was mediated by hADSC-inhibiting ROS, not by the stem cell engraftment.


Assuntos
Tecido Adiposo/citologia , Disfunção Cognitiva/terapia , Infarto do Miocárdio/terapia , Transplante de Células-Tronco , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Disfunção Cognitiva/fisiopatologia , Humanos , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Estresse Oxidativo , Ratos Wistar , Células-Tronco , Superóxidos/sangue , Superóxidos/metabolismo , Microglobulina beta-2/metabolismo
8.
J Cell Mol Med ; 24(21): 12272-12284, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33022900

RESUMO

Functional decline of stem cell transplantation in ageing hosts is well documented. The mechanism for this is poorly understood, although it is known that advancing age does not provide an optimal milieu for exogenous stem cells to survive, engraft and differentiate. We showed that n-butylidenephthalide improved human adipose-derived stem cell (hADSC) engraftment via attenuating the production of reactive oxygen species (ROS). It remained unclear whether pre-treated hosts with n-butylidenephthalide can rejuvenate the ageing heart and improve hADSC engraftment by regulating the ROS/NLRP3 inflammasome-mediated cardiac fibrosis after myocardial infarction. One hour after coronary ligation, hADSCs were transplanted into the hearts of young and ageing Wistar rats that were pre-treated with or without n-butylidenephthalide for 3 days. At day 3 after infarction, myocardial infarction was associated with an increase in ROS levels and NLRP3 inflammasome activity with age. hADSC transplant effectively provided a significant decrease in ROS levels, NLRP3 inflammasome activity, IL-1ß levels and cardiac fibrosis in either young or old infarcted rats. However, the beneficial effects of hADSCs were greater in young compared with old rats in terms of NLRP3 inflammasome activity. The infarcted ageing rats pre-conditioned by n-butylidenephthalide improved engraftment and differentiation of hADSCs and additionally attenuated cardiac fibrosis compared with hADSCs alone. The anti-inflammation effects of n-butylidenephthalide were reversed by SIN-1. In conclusions, the increased NLRP3 inflammasome activity plays the pathogenesis of ageing-related functional hADSC decline in the ageing hosts. n-butylidenephthalide-pre-treated ageing hosts reversibly ameliorate the harsh microenvironments, improve stem cell engraftment and attenuate cardiac fibrosis after myocardial infarction.


Assuntos
Tecido Adiposo/citologia , Envelhecimento , Inflamassomos/metabolismo , Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transplante de Células-Tronco , Animais , Diferenciação Celular , Fibrose , Hemodinâmica , Humanos , Interleucina-1beta/metabolismo , Masculino , Miocárdio/patologia , Proteínas de Neoplasias/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Fenótipo , Anidridos Ftálicos/farmacologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/citologia , Superóxidos/metabolismo
9.
Cell Transplant ; 29: 963689720927398, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32648485

RESUMO

Osteoporotic fracture is the main complication of osteoporosis (OP) and accounts for millions of injuries annually. Local intervention by intra-marrow injection has been a good option for preventing osteoporotic bone loss when the osteoporotic femoral fracture has been treated. In this study, tail vein transplantations were examined to evaluate the cell-based therapeutic approach for treating OP with adipose-derived stem cells (ADSCs) and platelet-rich fibrin releasates (PRFr) in an ovariectomized (OVX) mice model. Thirty-six 12-wk-old female ICR mice were randomly divided into six groups: untreated control; sham-operated; OVX-control; OVX-ADSCs; OVX-PRFr; and OVX-ADSCs+PRFr. Starting 8 wk after ovariectomy, the OVX mice received tail vein injections once each week for four consecutive weeks, then were evaluated radiographically and histopathologically 8 wk after the first injection. We also assessed changes to bone trabeculae in the proximal tibial growth plate. In OVX mice treated with ADSCs or PRFr alone, or with a combination of ADSCs and PRFr, the trabecular bone mineral density (BMD), bone volume ratios (BV/TV), and numbers (Tb.N) in the proximal tibia areas were significantly higher than that in the OVX-control group. Significant differences between OVX-treated mice and OVX controls were found for trabecular separation, but not for trabecular thickness. These results indicate that ADSCs or PRFr treatment enhances bone microarchitecture in OP. The treatment of bone loss of OVX mice with ADSCs+PRFr induced greater bone consolidation with bone tissue production (P < 0.01) when compared to the others. Thus, we conclude that the transplantation of ADSCs combined with PRFr might provide an alternative strategy for the treatment of various bone disorders in OP with an unlimited source of cells and releasates.


Assuntos
Tecido Adiposo/transplante , Osteoporose/cirurgia , Osteoporose/terapia , Fibrina Rica em Plaquetas/metabolismo , Transplante de Células-Tronco/métodos , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Coelhos
10.
Cell Transplant ; 29: 963689720919438, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32538130

RESUMO

Sciatic nerve injuries, not uncommon in trauma with a limited degree of functional recovery, are considered a persistent clinical, social, and economic problem worldwide. Accumulating evidence suggests that stem cells can promote the tissue regeneration through various mechanisms. The aim of the present study was to investigate the role of adipose tissue-derived stem cells (ADSCs) and combine with platelet-rich fibrin releasate (PRFr) in the regeneration of sciatic nerve injury in rats. Twenty-four Sprague-Dawley rats were randomly assigned to four groups, a blade was used to transect the left hindlimb sciatic nerve, and silicon tubes containing one of the following (by injection) were used to bridge the nerve proximal and distal ends (10-mm gap): group 1: untreated controls; group 2: PRFr alone; group 3: ADSCs alone; group 4: PRFr + ADSCs-treated. Walking function was assessed in horizontal rung ladder apparatus to compare the demands of the tasks and test sensitivity at 1-mo interval for a total of 3 mo. The gross inspection and histological examination was performed at 3 mo post transplantation. Overall, PRFr + ADSCs-treated performed better compared with PRFr or ADSCs injections alone. Significant group differences of neurological function were observed in ladder rung walking tests in all treated groups compared to that of untreated controls (P < 0.05). This injection approach may provide a successfully employed technique to target sciatic nerve defects in vivo, and the combined strategy of ADSCs with PRFr appears to have a superior effect on nerve repair.


Assuntos
Tecido Adiposo/fisiopatologia , Fibrina Rica em Plaquetas/metabolismo , Nervo Isquiático/fisiopatologia , Células-Tronco/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Ratos , Ratos Sprague-Dawley
11.
Cell Transplant ; 28(1_suppl): 100S-111S, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31722556

RESUMO

Currently, the only effective therapy for cirrhosis of the liver is liver transplantation. However, finding a compatible liver is difficult due to the low supply of healthy livers and the ever-increasing demand. However, stem-cell therapy may offer a solution for liver cirrhosis; for example, GXHPC1 therapy preparation contains adipose-derived mesenchymal stem cells (AD-MSCs) and was developed for the treatment of liver cirrhosis. In our previous report, animal studies suggested that treatment of a diseased liver via GXHPC1 transplantation can abrogate liver fibrosis and facilitate recovery of liver function. In our current human trial, patients with liver cirrhosis were included. Their adipose tissue was harvested from the subcutaneous fat of the abdominal wall during surgery. AD-MSCs were cultured and suspended at a concentration of 100 million cells in 1 ml of physiological saline (i.e., GXHPC1). This human study passed the Taiwan Food and Drug Administration IND inspection and received Phase I clinical trial permission. The trial was conducted with six patients with liver cirrhosis to demonstrate the safety and efficacy of administering GXHPC1. Intrahepatic injection of GXHPC1 did not cause any safety issues in the analysis of adverse drug reactions and suspected unexpected serious adverse reactions, and showed a tendency for improvement of liver function, METAVIR score, Child-Pugh score, MELD score, and quality of life for patients with liver cirrhosis.


Assuntos
Tecido Adiposo/citologia , Cirrose Hepática/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Tecido Adiposo/cirurgia , Adulto , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Cirrose Hepática/sangue , Cirrose Hepática/patologia , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Qualidade de Vida , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos
12.
Redox Biol ; 27: 101170, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31164286

RESUMO

Left ventricular hypertrophy (LVH) in hypertension has prognostic significance on cardiovascular mortality and morbidity. Recently, we have shown that n-butylidenephthalide (BP) improves human adipose-derived stem cell (hADSC) engraftment via attenuated reactive oxygen species (ROS) production. This prompted us to investigate whether remote transplantation of BP-pretreated hADSCs confers attenuated LVH at an established phase of hypertension. Male spontaneously hypertensive rats (SHRs) aged 12 weeks were randomly allocated to receive right hamstring injection of vehicle, clinical-grade hADSCs, and BP-preconditioned hADSCs for 8 weeks. As compared with untreated SHRs, naïve hADSCs decreased the ratio of LV weight to tibia, cardiomyocyte cell size, and collagen deposition independent of hemodynamic changes. These changes were accompanied by attenuated myocardial ROS production and increased p-STAT3 levels. Compared with naïve hADSCs, BP-preconditioned hADSCs provided a further decrease of ROS and LVH and an increase of local hADSC engraftment, STAT3 phosphorylation, STAT3 activity, STAT3 nuclear translocation, myocardial IL-10 levels, and the percentage of M2 macrophage infiltration. SIN-1 or S3I-201 reversed the effects of BP-preconditioned ADSCs increase on myocardial IL-10 levels. Furthermore, SIN-1 abolished the phosphorylation of STAT3, whereas superoxide levels were not affected following the inhibition of STAT3. Our results highlighted the feasibility of remote transplantation of hADSCs can be considered as an alternative procedure to reverse cardiac hypertrophy even at an established phase of hypertension. BP-pretreated hADSCs polarize macrophages into M2 immunoregulatory cells more efficiently than naïve hADSCs via ROS/STAT3 pathway.


Assuntos
Tecido Adiposo/fisiopatologia , Cardiomegalia/fisiopatologia , Hipertensão/fisiopatologia , Macrófagos/fisiologia , Células-Tronco/fisiologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos/fisiologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Cardiomegalia/metabolismo , Humanos , Hipertensão/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/fisiopatologia , Interleucina-10/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/fisiologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Anidridos Ftálicos/farmacologia , Ratos , Ratos Endogâmicos SHR , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
13.
Lab Invest ; 99(5): 634-647, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30683900

RESUMO

Stem cells can modify macrophage phenotypes; however, the mechanisms remain unclear. We investigated whether n-butylidenephthalide (BP) primed adipose-derived stem cells (ADSCs) attenuated cardiac fibrosis via regulating macrophage phenotype by a PI3K/STAT3-dependent pathway in postinfarcted rats. Male Wistar rats after coronary ligation were allocated to receive either intramyocardial injection of vehicle, ADSCs (1 × 106 cells), BP-preconditioned ADSCs, (BP + lithium)-preconditioned ADSCs, (BP + LY294002)-preconditioned ADSCs, and (BP + S3I-201)-preconditioned ADSCs. ADSCs were primed for 16 h before implantation. BP-pretreated ADSCs increased the cell viability compared with naive ADSCs in the in vitro experiments. Infarct sizes were similar among the infarcted groups at the acute and chronic stages of infarction. At day 3 after infarction, post-infarction was associated with increased M1 macrophage infiltration, which was inhibited by administering naive ADSCs. Compared with naive ADSCs, BP-preconditioned ADSCs provided a significant increase of Akt and STAT3 phosphorylation, STAT3 activity, STAT3 nuclear translocation, myocardial IL-10 levels, and the percentage of M2 macrophage infiltration. The effects of BP on M2 polarization were reversed by LY294002 or S3I-201. Furthermore, the phosphorylation of both Akt and STAT3 was abolished by LY294002, whereas Akt phosphorylation was not affected following the inhibition of STAT3. The addition of lithium did not have additional effects compared with BP alone. After 4 weeks of implantation, ADSCs remained in the myocardium, and reduced fibrosis and improved cardiac function. BP-preconditioned ADSCs provided superior cardioprotection, greater ADSC engraftment, and antifibrotic effects compared with naive ADSCs. These results suggest that BP-pretreated ADSCs polarize macrophages into M2 cells more efficiently than naive ADSCs via the PI3K/STAT3 pathway.


Assuntos
Adipócitos/citologia , Ativação de Macrófagos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Anidridos Ftálicos/farmacologia , Fator de Transcrição STAT3/metabolismo , Células-Tronco/efeitos dos fármacos , Animais , Cardiotônicos/farmacologia , Células Cultivadas , Cromonas/farmacologia , Fibrose/prevenção & controle , Humanos , Lítio/farmacologia , Masculino , Morfolinas/farmacologia , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/citologia , Células-Tronco/metabolismo
14.
Knee ; 25(6): 1181-1191, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30420268

RESUMO

BACKGROUND: This study aimed to investigate the efficacy of adipose-derived mesenchymal stem cells (ADSCs), platelet-rich fibrin releasates (PRFr), and chondrocyte transplantation in rabbit acute osteochondral defects. METHODS: Thirty rabbits were randomly assigned to five groups: untreated controls; ADSCs alone; PRFr alone; PRFr + ADSCs; and PRFr + chondrocytes. The critical size osteochondral defects in right knee femoral condyles were injected intra-articularly according to the groups, as listed. The experimental rabbits received treatments once a week for two weeks postoperatively. All evaluations were conducted for 14 weeks following surgery, and the regenerated cartilages were assessed by gross inspection and histological examination. RESULTS: There were no complications encountered in any of the rabbits. The size of the defect decreased and the volume of repaired cartilage increased in the medial femoral condyles of the PRFr + ADSCs group. Relative to the ADSCs or PRFr group, histological examination demonstrated that the PRFr + ADSCs group had thicker hyaline cartilage-specific extracellular matrix. Grading scores revealed that PRFr + ADSCs injection had better matrix, cell distribution, and surface indices than other groups (P < 0.05). However, the histological scores reported for PRFr + chondrocytes on cartilage repair were similar to those of PRFr, and there were no significant between-group differences. CONCLUSIONS: These findings showed that intra-articular injections of PRFr + ADSCs into the knee can reduce cartilage defects by regenerating hyaline-like cartilage without complications. This approach may provide an alternative method for functional reconstruction of acute osteochondral defects with an unlimited source of cells and releasates.


Assuntos
Cartilagem Articular/lesões , Cartilagem Articular/fisiologia , Condrócitos/transplante , Transplante de Células-Tronco Mesenquimais , Fibrina Rica em Plaquetas , Regeneração , Tecido Adiposo/citologia , Animais , Injeções Intra-Articulares , Modelos Animais , Coelhos
15.
J Mol Cell Cardiol ; 104: 17-30, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28130118

RESUMO

Adipose-derived stem cell (ADSC) transplantation is a promising new therapy to improve cardiac function after myocardial infarction. However, its low efficacy of transdifferentiation hampers its usefulness. Glycogen synthase kinase-3ß (GSK-3ß) signal has been shown to play a role in preconditioning-induced cardioprotection. We assessed whether n-butylidenephthalide (BP) primed ADSCs can attenuate arrhythmias by a GSK-3ß-dependent pathway after myocardial infarction. Male Wistar rats after coronary ligation was randomly allocated to receive intramyocardial injection of vehicle, ADSCs, BP-preconditioned ADSCs, (BP+lithium)-preconditioned ADSCs, (BP+SB216763)-preconditioned ADSCs, and (BP+LY294002)-preconditioned ADSCs. ADSCs were primed for 16h before implantation. After 4weeks of implantation, ADSCs were retained in myocardium, reduced fibrosis and improved cardiac function. Sympathetic hyperinnervation was blunted after administering ADSCs, assessed by immunofluorescent analysis, and Western blotting and real-time quantitative RT-PCR of nerve growth factor. Arrhythmic scores during programmed stimulation in the ADSC-treated infarcted rats were significantly lower than vehicle. BP-preconditioned ADSCs had superior cardioprotection, greater ADSC engraftment and transdifferentiation, and antiarrhythmic effects compared with ADSCs alone. Simultaneously, BP increased the levels of phospho-Akt and down-regulated GSK-3ß activity. The effects of BP against sympathetic hyperinnervation were blocked by LY294002, a PI3K inhibitor. Addition of either lithium or SB216763 did not have additional effects compared with BP alone. Compared with ADSC alone, BP-primed ADSC implantation improved stem cell engraftment and attenuated sympathetic hyperinnervation and arrhythmias through a PI3K/Akt/GSK-3ß-dependent pathway, suggesting that a synergic action was achieved between BP pretreatment and ADSCs.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/metabolismo , Arritmias Cardíacas/etiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Infarto do Miocárdio/complicações , Infarto do Miocárdio/metabolismo , Fator de Crescimento Neural/metabolismo , Transdução de Sinais , Células-Tronco Adultas/efeitos dos fármacos , Animais , Arritmias Cardíacas/diagnóstico , Biomarcadores , Transdiferenciação Celular/efeitos dos fármacos , Ecocardiografia , Fibrose , Testes de Função Cardíaca , Hemodinâmica , Humanos , Imuno-Histoquímica , Masculino , Infarto do Miocárdio/diagnóstico , Neovascularização Patológica , Fenótipo , Anidridos Ftálicos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transplante de Células-Tronco
16.
Cell Transplant ; 26(3): 449-460, 2017 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-27718343

RESUMO

Human adipose-derived stem cells (hADSCs) are a promising source of autologous stem cells for personalized cell-based therapies. Culture expansion of ADSCs provides an attractive opportunity for liver cirrhosis patients. However, safety and stability issues can pose big challenges for personalized autologous stem cell products. In the present study, we addressed whether the commercial production program could provide a consistent product for liver cirrhosis therapy. We collected adipose tissue from three human donors by lipoaspirate and isolated ADSCs, which were expanded in culture to reach 1 × 108 cells (an approximately 1,000-fold expansion) within four passages. We then examined their morphology, chromosome stability, surface markers, and differentiation ability after culture. Next, we explored their therapeutic potential using a rat model of thioacetamide-induced liver cirrhosis. Culture-expanded ADSCs were injected intrahepatically, and their biodistribution was tracked by immunohistochemistry using an antibody against human mitochondria. Finally, we tested for tumor development by subcutaneously injecting a 100-fold dose range of cultured ADSCs into immunocompromised mice. Taken together, we find that culture expansion of autologous ADSCs is a potentially suitable stem cell product for personalized cell-based therapy for patients with liver cirrhosis.


Assuntos
Cirrose Hepática/terapia , Células-Tronco/citologia , Adipogenia/genética , Adipogenia/fisiologia , Tecido Adiposo/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Condrogênese/genética , Condrogênese/fisiologia , Feminino , Humanos , Imuno-Histoquímica , Imunofenotipagem , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Osteogênese/genética , Osteogênese/fisiologia , Ratos , Transplante de Células-Tronco , Células-Tronco/fisiologia , Transplante Autólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA