Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Alcohol ; 114: 9-24, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37597575

RESUMO

Evidence has suggested that addiction and memory systems are related, but the signaling cascades underlying this interaction have not been completelyealed yet. The importance of calcium-calmodulin-dependent protein kinase II (CaMKII) and brain-derived neurotrophic factor (BDNF) in the memory processes and also in drug addiction has been previously established. In this present investigation, we examined the effects of repeated morphine pretreatment on impairment of spatial learning and memory acquisition induced by systemic ethanol administration in adult male rats. Also, we assessed how these drug exposures influence the expression level of CaMKII and BDNF in the hippocampus and amygdala. Animals were trained by a single training session of 8 trials, and a probe test containing a 60-s free-swim without a platform was administered 24 h later. Before training trials, rats were treated with a once-daily subcutaneous morphine injection for 3 days followed by a 5-day washout period. The results showed that pre-training ethanol (1 g/kg) impaired spatial learning and memory acquisition and down-regulated the mRNA expression of CaMKII and BDNF. The amnesic effect of ethanol was suppressed in morphine- (15 mg/kg/day) pretreated animals. Furthermore, the mRNA expression level of CaMKII and BDNF increased significantly following ethanol administration in morphine-pretreated rats. Conversely, this improvement in spatial memory acquisition was prevented by daily subcutaneous administration of naloxone (2 mg/kg) 15 min prior to morphine administration. Our findings suggest that sub-chronic morphine treatment reverses ethanol-induced spatial memory impairment, which could be explained by modulating CaMKII and BDNF mRNA expressions in the hippocampus and amygdala.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Morfina , Ratos , Masculino , Animais , Morfina/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Memória Espacial , Hipocampo , RNA Mensageiro/metabolismo
2.
Appl Biochem Biotechnol ; 195(8): 4796-4817, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37184724

RESUMO

Islet transplantation offers improved glycemic control in individuals with type 1 diabetes mellitus. However, in vitro islet culture is associated with islet apoptosis and eventually will lose their functionality prior to transplantation. In this study, we examined the effects of mesenchymal stem cells (MSCs) secretome preconditioned with diazoxide (DZ) and trimetazidine (TMZ) on rat islet cells during pre-transplant culture. With and without preconditioned hAD-MSCs' concentrated conditioned media (CCM) were added to the culture medium containing rat islets every 12 h for 24 and 48 h, after testing for selected cytokine concentrations (interleukin (IL)-4, IL-6, IL-13). Insulin content, glucose-stimulated insulin secretion, islet cell apoptosis, and mRNA expression of pro-apoptotic (BAX, BAK-1, and PUMA) and anti-apoptotic factors (BCL-2, BCL-xL, and XIAP) in rat islets were assessed after 24 and 48 h of culture. The protein level of IL-6 and IL-4 was significantly higher in TMZ-MSC-CM compared to MSC-non-CM. In rat isolated islets, normalized secreted insulin in the presence of 16.7 mM glucose was significantly higher in treated islet groups compared to control islets at both 24 and 48 h cultivation. Also, the percentage of apoptotic islet cells TMZ-MSC-CCM-treated islets was significantly lower compared to MSC-CM and MSC-CCM-treated islets in both 24 and 48 h cultivation. Consistent with the number of apoptotic cells, after 24 h culture, the expression of BCL-2 and BCL-xL genes in the control islets was lower than all treatment islet groups and in 48 h was lower than only TMZ-MSC-CM-treated islets. Also, the expression of the XIAP gene in control islets was significantly lower compared to the TMZ-MSC-CCM-treated islets at both at 24 and 48 h. In addition, mRNA level of the BAX gene in TMZ-MSC-CCM-treated islets was significantly lower compared to other groups at 48 h. Our findings revealed that TMZ proved to be more effective than DZ and could enhance the potential of hAD-MSCs-CM to improve the function and viability of islets prior to transplantation.


Assuntos
Ilhotas Pancreáticas , Células-Tronco Mesenquimais , Trimetazidina , Ratos , Animais , Trimetazidina/farmacologia , Trimetazidina/metabolismo , Interleucina-6/metabolismo , Secretoma , Proteína X Associada a bcl-2/metabolismo , Ilhotas Pancreáticas/metabolismo , Insulina/metabolismo , Diazóxido/metabolismo , Diazóxido/farmacologia , Glucose/metabolismo
3.
CNS Neurosci Ther ; 28(9): 1425-1438, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35715988

RESUMO

AIMS: Regenerative medicine literature has demonstrated that the therapeutic potentials of mesenchymal stem cells (MSCs) in experimental stroke are attributed to secreted bioactive factors rather than to cell replacement. Here, we explored the effects of secretome or conditioned medium (CM) derived from human embryonic stem cell-derived MSCs (hESC-MSCs) on hippocampal neurogenesis, inflammation, and apoptosis in experimental stroke. METHODS: Ischemic stroke was induced by right middle cerebral artery occlusion (MCAO) in male Wistar rats, and CM was infused either one time (1-h post-stroke; CM1) or three times (1-, 24-, and 48-h post-stroke; CM3) into left lateral ventricle. Neurogenesis markers (Nestin, Ki67, Doublecortin, and Reelin) were assessed at transcript and protein levels in the dentate gyrus of the hippocampus on day seven following MCAO. In parallel, changes in the gene expression of markers of apoptosis (Bax and Bim, as well as an anti-apoptotic marker of Bcl2), inflammation (IL-1ß and IL-6, as well as IL-10 as an anti-inflammatory cytokine), trophic factors (BDNF, GDNF, NGF, and NT-3), and angiogenesis (CD31 and VEGF) in the hippocampus were assessed. RESULTS: Our results demonstrate that CM3 treatment could stimulate neurogenesis and angiogenesis concomitant with inhibition of inflammation, apoptosis, and neuronal loss in ischemic brains. Furthermore, rats treated with CM3 exhibited upregulation in neurotrophic factors. CONCLUSION: Our results suggest that hESC-MSC-CM could promote neurogenesis and protect brain tissue from ischemic injury, partly mediated by induction of angiogenesis and neurotrophic factors and inhibition of inflammatory and apoptotic factors expression.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Acidente Vascular Cerebral , Animais , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Humanos , Infarto da Artéria Cerebral Média/complicações , Inflamação/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Fatores de Crescimento Neural/metabolismo , Neurogênese , Neuroproteção , Ratos , Ratos Wistar , Secretoma , Acidente Vascular Cerebral/metabolismo
4.
Biomed Pharmacother ; 140: 111709, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34020250

RESUMO

It is well accepted that the success of mesenchymal stem cells (MSCs) therapy against experimental stroke is mainly due to cellular paracrine manners rather than to replace lost tissue per se. Given such "bystander" effects, cell-free therapeutics manifest as a promising approach in regenerative medicine. Here we aimed at evaluating the effect of conditioned medium (CM) derived from human embryonic MSCs (hESC-MSC) on the neurological deficit, neurogenesis, and angiogenesis in experimental stroke. Adult male Wistar rats subjected to middle cerebral artery occlusion (MCAO), were treated with intracerebroventricular CM either one time (1 h post MCAO) or three times (1, 24, and 48 h post MCAO). Motor performance was assessed by the cylinder test on days 3 and 7. Cerebral samples were obtained for infarct size and molecular analysis on day 7 post-injury. Neurogenesis was evaluated by probing Nestin, Ki67, DCX, and Reelin transcripts and protein levels in the striatum, cortex, subventricular zone, and corpus callosum. The mRNA and protein expression of CD31 were also assessed in the striatum and cortical region to estimate angiogenesis post MCAO. Our findings demonstrate that CM treatment could significantly ameliorate neurological deficits and infarct volume in MCAO rats. Furthermore, ischemic stroke was associated with higher levels of neurogenesis and angiogenesis markers. Following treatment with CM, these markers were further potentiated in the brain regions. This study suggests that the therapeutic benefits of CM obtained from hESC-MSCs at least partly are mediated through improved neurogenesis and angiogenesis to accelerate the recovery of cerebral ischemia insult.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Células-Tronco Embrionárias Humanas/citologia , Infarto da Artéria Cerebral Média/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Células-Tronco Mesenquimais , Neovascularização Fisiológica/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Proteína Duplacortina , Humanos , Infarto da Artéria Cerebral Média/fisiopatologia , Injeções Intraventriculares , AVC Isquêmico/fisiopatologia , Masculino , Ratos Wistar , Proteína Reelina
5.
J Cell Physiol ; 236(3): 1967-1979, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32730642

RESUMO

The transplantation of mesenchymal stem cells (MSCs) is of main approaches in regenerative therapy for stroke. Due to the potential tumorigenicity and low survival rate of transplanted cells, focuses have been shifted from cell replacement to their paracrine effects. Therefore, stem cell-conditioned medium (CM) therapy has emerged as an alternative candidate. Here, we investigated the effect of CM derived from human embryonic MSCs on experimental ischemic stroke. Wistar rats underwent ischemic stroke by the right middle cerebral artery occlusion (MCAO). CM was infused either one time (1 hr post-MCAO) or three times (1, 24, and 48 hr post-MCAO) through guide cannula into the left lateral ventricle. Neurological functions were evaluated using Bederson's test and modified Neurological Severity Score on Days 1, 3, and 7 following MCAO. Infarction volumes and cerebral edema were measured on Days 3 and 7. growth-associated protein-43, synaptophysin, cAMP response element-binding protein, and phosphorylated-cAMP response element-binding protein levels were also assessed in peri-ischemic cortical tissue on Day 7 postsurgery. Our results indicated that three times injections of CM could significantly reduce body weight loss, mortality rate, infarct volumes, cerebral edema, and improve neurological deficits in MCAO rats. Moreover, three injections of CM could restore decreased levels of synaptic markers in MCAO rats up to its normal levels observed in the sham group. Our data suggest that using the CM obtained from embryonic stem cells-MSCs could be a potent therapeutic approach to attenuate cerebral ischemia insults which may be partly mediated through modulation of synaptic plasticity.


Assuntos
Encéfalo/patologia , Meios de Cultivo Condicionados/farmacologia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Mesenquimais/metabolismo , Acidente Vascular Cerebral/patologia , Sinapses/patologia , Animais , Biomarcadores/metabolismo , Peso Corporal/efeitos dos fármacos , Infarto Encefálico/complicações , Infarto Encefálico/patologia , Linhagem Celular , Edema/complicações , Edema/patologia , Humanos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Injeções Intraventriculares , Masculino , Neurogênese/efeitos dos fármacos , Ratos Wistar , Sinapses/efeitos dos fármacos
6.
Mol Biol Rep ; 47(3): 1859-1869, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32016633

RESUMO

Cancer stem cells are commonly tolerant toward chemotherapy and radiotherapy. Oct4 and Sox2 transcription factors are shown to be overexpressed in various cancers. At the current research, inhibition of Oct4 and Sox2 transcription factors was performed through application of decoy oligodeoxynucleotides (ODNs) strategy via repressing stemness properties in HT29-ShE cells encompassing enriched cancer stem-like cells. Designed Oct4-Sox2 complex decoy ODNs were transfected into HT29-ShE cells with Lipofectamine reagent. At the next step, ODNs efficiency transfection and subcellular localization were determined via flow cytometry and fluorescence microscopy, respectively. Further investigations such as cell proliferation and apoptosis analysis, colonosphere formation, invasion and migration, and real-time PCR assays were also carried out. Obtained results shed light on the fact that the designed complex decoys were effectively transfected into HT29-ShE cells, and they were found to be localized in subcellular compartments. Oct4-Sox2 decoy ODNs led to decreased cell viability, arresting the cell cycle in G0/G1 phases, increasing apoptosis, inhibition of migration/invasion and colonosphere formation ability of HT29-ShE cells in comparison with control and scramble groups. Furthermore, Oct4-Sox2 complex decoy could modulate the MET process via alteration of mRNA expression of downstream genes. It could be concluded that application of Oct4-Sox2 transcription factor decoy strategy in cells with stemness potential could lead to inhibiting the cell growth and triggering differentiation. Therefore, this technique could be applied along with usual remedies (chemotherapy and radiotherapy) as high potential method for treating cancer.


Assuntos
Células-Tronco Neoplásicas/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Oligodesoxirribonucleotídeos/farmacologia , Fatores de Transcrição SOXB1/genética , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Células HT29 , Humanos , Microscopia de Fluorescência , Complexos Multiproteicos/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo
7.
J Cell Physiol ; 235(6): 5429-5444, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31912904

RESUMO

Due to the presence of cancer stem cells (CSCs), breast cancer often relapsed after conventional therapies. Strategies that induce differentiation of CSCs will be helpful in eradication of tumor cells, so we designed an oligodeoxynucleotide (ODNs) for targeting of signal transducer and activator of transcription 3 (STAT3) transcription factor which is involved in stemness, and constitutively activated in triple-negative breast cancer. Molecular docking and electrophoretic mobility shift assay analysis showed that decoy ODN bound specifically to the DNA binding site of STAT3 protein. The prevalent uptake of Cy3-labeled ODNs is in the cytoplasm and the nucleus of MDA-MB-231 treated cells. STAT3 decoy ODNs treatment showed cell growth inhibition by decreasing cell viability (17%), increasing the percentage of arrested cells in G0/G1 phases (18%), and triggering apoptosis (29%). Migration and invasion potential decreased from 10.77 to 6.76 µm/hr, by wound closure rate, and migrated/invaded percentage by 26.4% and 15.4% in the transwell assays, respectively. CD44 protein expression level on the cell surface also decreased, while CD24 increased. Mammosphere formation efficiency reduced in terms of tumorsphere size by 47%, while the required time increased. Cells morphology was changed, and lipid droplets were accumulated in the cytoplasm compared to the control and scrambled groups, in all assays (repeated triplicate). Furthermore, the gene expression of all downstream targets significantly decreased owing to suppressing the STAT3 transcription factor. Overall, the results confirmed the antitumor effects of STAT3 decoy in MDA-MB-231 cells. Thus, it seems that STAT3 decoy ODNs might be considered as an auxiliary tool for breast cancer eradicating by the differentiation therapy approach.


Assuntos
Neoplasias da Mama/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Oligodesoxirribonucleotídeos/farmacologia , Fator de Transcrição STAT3/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Simulação de Acoplamento Molecular , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Oligodesoxirribonucleotídeos/química , Proteólise , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/química
8.
Cell Biol Int ; 44(1): 14-26, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31393053

RESUMO

Mesenchymal stem cells (MSCs), which are known for having therapeutic applications, reside in stem cell niches where the oxygen concentration is low. At the molecular level, the master regulator of the cellular reaction to hypoxia is hypoxia-inducible transcription factor (HIF). The transcriptional response of a cell to hypoxia is affected by two major components; first, the structure of hypoxia-response elements (HREs), which primarily define how much of the HIF signal is integrated into the transcriptional output of individual genes. Second, the availability of other transcriptional factors cooperating with HIF in the context of HRE. In MSCs, the expression of a single gene by hypoxia depends on elements such as factors influencing the HIF activity, metabolic pathways, the real oxygen concentration in the cellular microenvironment, and duration of culture. In addition, specific growth factors and pro-infection cytokines, hormones, oncogenic signaling, as well as ultrasound are potent regulators of HIF in MSCs. Altogether, the response of MSCs to hypoxia is complex and mediated by several genes and molecular agents. Regarding the influence of hypoxia on MSCs, oxygen concentration must be taken into consideration based on the cell type and the aim of culture before a particular MSCs culture.

9.
J Cell Physiol ; 234(12): 22935-22946, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31111504

RESUMO

A better understanding of cancer stem cells (CSCs) may facilitate the prevention and treatment of cancers. Epithelial-mesenchymal transition (EMT) is a process activated during invasion and metastasis of tumors. EMT induction in normal and tumor cells makes them more resistant to chemotherapy. E-cadherin is a membrane protein and plays a role in tumor invasion, metastasis, and prognosis. Downregulation of E-cadherin is a hallmark of EMT. Here, we created a model of cancer stem-like cells enrichment via EMT induction using E-cadherin downregulation in HT29 cell line using a lentiviral vector carrying shRNA. We aimed to evaluate cancer and anti-CSC chemotherapeutics screening. The markers of EMT and CSCs were assessed and compared with control cells using flow cytometry, real-time PCR, immunocytochemistry, western blot, migration assay, invasion assay, and colony formation assay. The transduced cells showed a mesenchymal morphology. High levels of EMT-related proteins were also expressed. These results confirmed that the transduced cells underwent EMT. In addition, we observed an increased population of E-cadherin-downregulated HT29 cell line among the cells expressing colon CSC markers (CD133+ and CD44+ ) after EMT induction. E-cadherin-downregulated cells were morphologically like mesenchymal cells, and the number of CD133+ - and CD44+ -cells (CSC-like cells) increased. These cells can be used as stable models to study cancer cells and screening of antitumor therapeutics.


Assuntos
Caderinas/genética , Neoplasias do Colo/genética , Transição Epitelial-Mesenquimal/genética , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133/genética , Caderinas/antagonistas & inibidores , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Vetores Genéticos/genética , Células HT29 , Humanos , Receptores de Hialuronatos/genética , Lentivirus/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Células-Tronco Neoplásicas/patologia , RNA Interferente Pequeno/genética
10.
J Cell Physiol ; 234(12): 22765-22774, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31115051

RESUMO

Expression of master transcriptional regulators of stem cells (Oct4 and Sox2) is associated with mediating tumor proliferation and tumor differentiation. The main goal of this study is the investigation of specific binding of designed Oct4-Sox2 transcription factors decoy oligodeoxynucleotides (ODNs) sequence to their nucleus-extracted proteins in HT29-ShE cells containing enriched cancer stem-like cells (SCLCs). First, gene expression of Oct4, Sox2, and E-cadherin revealed the overexpression of Oct4 and Sox2 and downregulation of E-cadherin in HT29-ShE cells compared with HT29 wild-type and HT29-ShC cells. Next, Oct4-Sox2 complex decoy ODNs were designed according to their elements in the promoter region of Sox2 gene. Then, the interactions of Oct4 and Sox2 proteins to designed ODNs were evaluated in silico. Finally, DNA-protein interactions of decoy ODNs and their corresponding proteins were examined by electrophoretic mobility shift assay (EMSA). Analysis of gel shift retardation assay admitted the specific binding of designed ODNs sequence to the nuclear extracted Oct4 and Sox2 proteins. The results will be a promising approach to target cancer stem cells for potential use in differentiation therapy before chemotherapy and radiotherapy of cancers.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator 3 de Transcrição de Octâmero/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Fatores de Transcrição SOXB1/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/metabolismo , Sítios de Ligação , Caderinas/genética , Caderinas/metabolismo , Forma Celular/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Desenho de Fármacos , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Simulação de Acoplamento Molecular , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator 3 de Transcrição de Octâmero/genética , Oligodesoxirribonucleotídeos/genética , Oligodesoxirribonucleotídeos/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/genética , Transdução de Sinais
11.
Cell Biol Int ; 43(8): 852-862, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31033094

RESUMO

The transcription factor T-cell factor 3 (TCF3), one component of the Wnt pathway, is known as a cell-intrinsic inhibitor of many pluripotency genes in embryonic stem cells (ESCs) that influences the balance between pluripotency and differentiation. In this study, the effects of inhibition of TCF3 transcription factor on the stemness of mouse ESCs (mESCs) were investigated using the decoy oligodeoxynucleotides (ODNs) strategy. The TCF3 decoy and its scramble ODNs were designed and synthesized. The interaction specificity of the TCF3 decoy with the TCF3 transcription factor was evaluated by the electrophoretic mobility shift assay. Subcellular localization was carried out using fluorescence and confocal microscopy. Self-renewal and pluripotency of mESCs were analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), cell cycle and apoptosis, alkaline phosphatase (ALP), embryoid body (EB) formation, and real-time assays. All experiments were performed in triplicate. The results showed that knockdown of TCF3 by decoy ODNs transfection in mESCs led to an increase in the cell proliferation, ALP enzyme activity, and master regulatory stemness genes and a decrease in the number and diameter of EBs. These results supported TCF3 as a potential target to maintain the pluripotency and self-renewal capacity of mESCs. Knockdown of the TCF3 transcription factor using decoy ODNs can be a promising method to maintain the stemness of stem cells in regenerative medicine and cell therapy researches.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Medicina Regenerativa , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Oligodesoxirribonucleotídeos/genética , Via de Sinalização Wnt/genética
12.
Artif Cells Nanomed Biotechnol ; 46(sup2): 1178-1187, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29929400

RESUMO

Systemic inflammatory response syndrome is a complex pathophysiologic and immunologic response to an insult. Sepsis is a life-threatening condition happening when the body's response to infection causes injury to its own tissues and organs. Stem cell therapy is a new approach to modulate immune responses. Mesenchymal stem cells (MSCs) establish a regenerative niche by secreting secretome and modulating immune responses. MSC secretome can be leveraged for therapeutic applications if production of secretary molecules were optimized. Pharmacological preconditioning using small molecules can increase survival of MSCs after transplantation. The aim of this study was to investigate the effect of secretome of human embryonic-derived mesenchymal stem cells (hESC-MSCs) preconditioned with MG-132,Trimetazidine (TMZ) and Diazoxide (DZ) on immunomodulatory efficiency of these cells in Lipo polysaccharide (LPS) challenged mice models. Mice were injected intraperitoneally with LPS and groups of animals were intraperitoneally given 1 ml 30× secretome 6 h after LPS injection. Serum levels of biochemical parameters were then measured by an auto analyser and serum inflammatory cytokine levels were analysed using commercially available RayBio Mouse Inflammation Antibody Array. Ultimately, histopathology and survival studies were conducted. The results showed that TMZ and DZ-conditioned medium significantly increasing the survival and improvement of histopathological score. We found that MG-132-conditioned medium failed to show significant outcomes. This study demonstrated that human MSC secretome has the potential to control inflammation.


Assuntos
Diazóxido/farmacologia , Células-Tronco Embrionárias Humanas/citologia , Leupeptinas/farmacologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Síndrome de Resposta Inflamatória Sistêmica/terapia , Trimetazidina/farmacologia , Animais , Linhagem Celular , Meios de Cultivo Condicionados/química , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Rim/metabolismo , Rim/patologia , Lipopolissacarídeos/farmacologia , Pulmão/metabolismo , Pulmão/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Análise de Sobrevida , Síndrome de Resposta Inflamatória Sistêmica/metabolismo , Síndrome de Resposta Inflamatória Sistêmica/patologia
13.
Oncol Lett ; 15(5): 8157-8164, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29849810

RESUMO

Over the latest decade, the role of microRNAs (miRNAs/miRs) has received more attention. miRNAs are small non-coding RNAs that may serve a role as oncogenes or tumor suppressor genes. Certain miRNAs regulate the apoptosis pathway by influencing pro- or anti-apoptotic genes. We hypothesized that increases in the expression of B cell lymphoma 2 (BCL2) and BCL2-like 1 (BCL2L1) genes, which have been reported in various types of cancer tissues, may be due to the downregulation of certain miRNAs. The present study aimed to identify miRNAs that target BCL2 and BCL2L1 anti-apoptotic genes in prostate cancer (PCa) clinical tissue samples. Certain candidate miRNAs were selected bioinformatically and their expression in PCa samples was analyzed and compared with that in benign prostatic hyperplasia (BPH) tissue samples. The candidate miRNAs that targeted BCL2 and BCL2L1 genes were searched in online databases (miRWalk, microRNA.org, miRDB and TargetScan). A total of 12 miRNAs that target the 3'-untranslated region of the aforementioned genes and/or for which downregulation of their expression has previously been reported in cancer tissues. A total of 30 tumor tissue samples from patients with PCa and 30 samples tissues from patients with BPH were obtained and were subjected to reverse transcription-quantitative polymerase chain reaction for expression analysis of 12 candidate miRNAs, and the BCL2 and BCL2L1 genes. Additionally, expression of 3 finally selected miRNAs and genes was evaluated in prostate cancer PC3 and DU145 cell lines and human umbilical vein endothelial cells. Among 12 miRNA candidates, the expression of miR-1266, miR-185 and miR-30c-2 was markedly downregulated in PCa tumor tissues and cell lines. Furthermore, downregulation of these miRNAs was associated with upregulation of the BCL2 and BCL2L1 genes. An inverse association between three miRNAs (miR-1266, miR-185 and miR-30c-2) and two anti-apoptotic genes (BCL2 and BCL2L1) may be considered for interventional miRNA therapy of PCa.

14.
Iran Biomed J ; 22(4): 237-45, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29132205

RESUMO

Background: Bone marrow mesenchymal stem cells (BM-MSCs) have emerged as a potential therapy for various inflammatory diseases. Because of some limitations, several recent studies have suggested the use of embryonic stem cell-derived MSCs (ESC-MSCs) as an alternative for BM-MSCs. Some of the therapeutic effects of the ESC-MSCs are related to the secretion of a broad array of cytokines and growth factors, known as secretome. Harnessing this secretome for therapeutic applications requires the optimization of production of secretary molecules. It has been shown that aggregation of MSCs into 3D spheroids, as a preconditioning strategy, can enhance immunomodulatory potential of such cells. In this study, we investigated the effect of secretome derived from human ESC-MSCs (hESC-MSCs) spheroids on secretion of IL-1ß, IL-10, and tumor necrosis factor α (TNF-α) from lipopolysaccharide (LPS)-induced peripheral blood mononuclear cells (PBMCs). Methods: In the present study, after immunophenotyping and considering mesodermal differentiation of hESC-MSCs, the cells were non-adherently grown to prepare 3D aggregates, and then conditioned medium or secretome was extracted from the cultures. Afterwards, the anti-inflammatory effects of the secretome were assessed in an in vitro model of inflammation. Results: Results from this study showed that aggregate-prepared secretome from hESC-MSCs was able to significantly decrease the secretion of TNF-α (301.7 ± 5.906, p < 0.0001) and IL-1ß (485.2 ± 48.38, p < 0.001) from LPS-induced PBMCs as the indicators of inflammation, in comparison with adherent culture-prepared secretome (TNF-α: 166.6 ± 8.04, IL-1ß: 125.2 ± 2.73). Conclusion: Our study indicated that cell aggregation can be an appropriate strategy to increase immunomodulatory characteristics of hESC-MSCs.

15.
Tissue Cell ; 49(5): 597-602, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28843336

RESUMO

Comprehensive proteome profiling of the factors secreted by mesenchymal stem cells (MSCs), referred to as secretome, revealed that it consists of cytokines, chemokines, growth factors, extracellular matrix proteins, and components of regeneration, vascularization, and hematopoiesis pathways. Harnessing this MSC secretome for therapeutic applications requires the optimization of production of secretary molecules. A variety of preconditioning methods have been introduced, which subject cells to stimulatory molecules to create the preferred response and stimulate persistent effects. Pharmacological preconditioning uses small molecules and drugs to increase survival of MSCs after transplantation or prolong release of effective secretary factors such as cytokines that improve immune system responses. In this study, we investigated the effect of secretome of human embryonic-derived mesenchymal stem cells (hESC-MSCs) preconditioned with Trimetazidine (TMZ) and Diazoxide (DZ) on immunomodulatory efficiency of these cells in LPS-induced peripheral blood mononuclear cells (PBMCs). PBMCs were isolated from human peripheral blood and treated with concentrated hESC-MSC-derived conditioned medium and then, the secreted levels of IL-10, TNFα and IL-1ß were assessed by ELISA after induction with LPS. The results showed that TMZ and DZ-conditioned medium significantly enhanced immunomodulatory potential of hESC-MSCs by increasing the secretion of IL-10, TNFα and IL-1ß from LPS- induced PBMCs. We also found that hESC-MSCs did not secrete mentioned cytokines prior to or after the preconditioning with TMZ and DZ. In conclusion, our results implied that TMZ and DZ can be used to promote the immunomodulatory effects of hESC-MSC secretome. It is obvious that for applying of these findings in clinical demands, the potency of different pre-conditioned MSCs secretome on immune response needs to be more clarified.


Assuntos
Diazóxido/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteoma/efeitos dos fármacos , Trimetazidina/farmacologia , Meios de Cultivo Condicionados , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Leucócitos Mononucleares/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Fator de Necrose Tumoral alfa/metabolismo , Vasodilatadores/farmacologia
16.
Anticancer Agents Med Chem ; 17(13): 1786-1795, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28403778

RESUMO

BACKGROUND: Myc (c-Myc) alone activates the embryonic stem cell-like transcriptional module in both normal and transformed cells. Its dysregulation might lead to increased cancer stem cells (CSCs) population in some tumor cells. OBJECTIVE: In order to investigate the potential of Myc decoy oligodeoxynucleotides for differentiation therapy, mouse embryonic stem cells (mESCs) were used in this study as a model of CSCs. To our best of knowledge this is the first report outlining the application of Myc decoy in transcription factor decoy "TFD" strategy for inducing differentiation in mESCs. METHODS: A 20-mer double-stranded Myc transcription factor decoy and scrambled oligodeoxynucleotides (ODNs) were designed, analyzed by electrophoretic mobility shift (EMSA) assay and transfected into the mESCs under 2 inhibitors (2i) condition. Further investigations were carried out using fluorescence and confocal microscopy, cell proliferation and apoptosis analysis, alkaline phosphatase and embryoid body formation assay, real-time PCR and western blotting. RESULTS: EMSA data showed that Myc decoy ODNs bound specifically to c-Myc protein. They were found to be localized in both cytoplasm and nucleus of mESCs. Our results revealed the potential capability of Myc decoy ODNs to decrease cell viability by (16.1±2%), to increase the number of cells arrested in G0/G1 phases and apoptosis by (14.2±3.1%) and (12.1±3.2%), respectively regarding the controls. Myc decoy could also modulate differentiation in mESCs despite the presence of 2i/LIF in our medium the presence of 2i/LIF in our medium. CONCLUSION: The optimized Myc decoy ODNs approach might be considered as a promising alternative strategy for differentiation therapy investigations.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Genes myc , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Oligodesoxirribonucleotídeos/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Meios de Cultura , Ensaio de Desvio de Mobilidade Eletroforética , Camundongos , Modelos Biológicos , Células-Tronco Embrionárias Murinas/enzimologia , Células-Tronco Neoplásicas/patologia , Reação em Cadeia da Polimerase em Tempo Real
17.
Iran Biomed J ; 21(1): 24-31, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27132108

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) are important candidates for MSC-based cellular therapy. Current paradigm states that MSCs support local progenitor cells in damaged tissue through paracrine signaling. Therefore, study of paracrine effects and secretome of MSCs could lead to the appreciation of mechanisms and molecules associated with the therapeutic effects of these cells. This study analyzed anti-inflammatory and immune-modulatory effects of MSC secretomes derived from embryonic stem cells (ESCs) and bone marrow cells after hypoxia and normoxia preconditioning. METHODS: ESCs differentiated into MSCs and characterized by flow cytometry and differentiation into adipocytes and osteoblasts. The experimental groups consisted of individual groups of ESC-MSCs and BM-MSCs (bone marrow-derived mesenchymal stromal cells), which were preconditioned with either hypoxia or normoxia for 24, 48 and 72 h. After collecting the cell-free medium from each treatment, secretomes were concentrated by centrifugal filters. Using a peripheral blood mononuclear cell (PBMC) assay and ELISA, IL-10 concentration in PBMCs was evaluated after their incubation with different secretomes from preconditioned and non-preconditioned MSCs. RESULTS: A significant difference was observed between ESC-MSC normoxia and ESC-MSC hypoxia in IL-10 concentration, and normoxia secretomes increased IL-10 secretion from PBMCs. Moreover, the strongest IL-10 secretion from PBMCs could be detected after the stimulation by ESC-MSC conditioned secretomes, but not BM-MSC conditioned medium. CONCLUSIONS: Human hypoxia preconditioned ESC-MSC secretome indicated stronger immune-modulatory effects compared to BM-MSC conditioned medium. It could be suggested that induced MSCs confer less immune-modulatory effects but produce more inflammatory molecules such as tumor necrosis factor α, which needs further investigation.

18.
Int J Artif Organs ; 39(10): 524-533, 2016 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-27901555

RESUMO

INTRODUCTION: Amid the plethora of methods to repair critical bone defects, there is no one perfect approach. In this study, we sought to evaluate a potent 3-dimensional (3D) bioactive SiO2-CaO-P2O5 glasses (bioglass)/gelatin (gel) scaffold for its biocompatibility by seeding cells as well as for its regenerative properties by animal implantation. METHODS: Osteoblast cells were seeded onto nanocomposite scaffolds to investigate the process of critical-size calvarial defect via new bone formation. Scanning electron microscopy (SEM) was used to validate topography of the scaffolds, its homogeneity and ideal cellular attachment. Proliferation assay and confocal microscopy were used to evaluate its biocompatibility. To validate osteogenesis of the bioactive nanocomposite scaffolds, they were first implanted into rats and later removed and analyzed at different time points post mortem using histological, immunohistochemical and histomorphometric methods. RESULTS: Based on in vitro results, we showed that our nanocomposite is highly cell-compatible material and allows for osteoblasts to adhere, spread and proliferate. In vivo results indicate that our nanocomposite provides a significant contribution to bone regeneration and is highly biodegradable and biocompatible. So, seeded scaffolds with osteoblasts enhanced repair of critical bone defects via osteogenesis. CONCLUSIONS: We demonstrate the feasibility of engineering a nanocomposite scaffold with an architecture resembling the human bone, and provide proof-of-concept validation for our scaffold using a rat animal model.


Assuntos
Substitutos Ósseos , Cerâmica , Osteoblastos , Crânio/cirurgia , Alicerces Teciduais , Animais , Regeneração Óssea , Adesão Celular , Linhagem Celular , Sobrevivência Celular , Gelatina/química , Teste de Materiais , Nanocompostos , Osteogênese , Ratos Wistar , Crânio/lesões , Cicatrização
19.
Neuropeptides ; 60: 13-19, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27751532

RESUMO

Behavioral abnormalities associated with opiate addiction include memory and learning deficits, which are the result of some alterations in the neuromodulatory systems. Recently, orexin has shown to influence drug addiction neural circuitry, specifically in mediating reward-related perception and memory. To explore the possible interaction of orexinergic and opioidergic system on modulation of learning and memory, we have investigated the effects of intra-dorsal hippocampal (intra-CA1) administration of orexin-1 receptor agonist and the competitive orexin-1 antagonist, SB-334867, on morphine-induced memory impairment by using step-down passive avoidance task in mice. Pre-training injection of morphine (5mg/kg, i.p.) impaired memory, which was restored when 24h later the same dose of the drug was administered. Pre-test administration of orexin-1 (0.5, 5 and 50pmol, intra-CA1) had not a significant effect on the retention latency compared to the saline-treated animals, but it restored the memory impairment induced by pre-training morphine (5mg/kg, i.p.). Pre-test administration of SB-334867 (10, 20 and 40nmol, intra-CA1) by itself decreased the retention latencies of passive avoidance task. Co-administration of orexin-1 (0.5, 5 and 50pmol, intra-CA1) and morphine (1mg/kg, i.p.) on the test day induced morphine state-dependent memory. Conversely, pre-test injection of SB-334867 (10, 20 and 40nmol, intra-CA1) inhibited the orexin-1-induced potentiation of morphine state-dependent learning on the test day. It is concluded that dorsal hippocampal orexin-1 receptors may be involved, at least in part, in morphine state-dependent learning in mice.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Memória/efeitos dos fármacos , Morfina/farmacologia , Entorpecentes/farmacologia , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Orexinas/farmacologia , Animais , Benzoxazóis/farmacologia , Hipocampo/metabolismo , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Naftiridinas , Ureia/análogos & derivados , Ureia/farmacologia
20.
Stem Cells Dev ; 25(24): 1898-1908, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27676103

RESUMO

Adult tissue-derived mesenchymal stem cells (MSCs) show tremendous promise for a wide array of therapeutic applications predominantly through paracrine activity. Recent reports showed that human embryonic stem cell (ESC)-derived MSCs are an alternative for regenerative cellular therapy due to manufacturing large quantities of MSCs from a single donor. However, no study has been reported to uncover the secretome of human ESC-MSCs as treatment of an acute liver failure (ALF) mouse model. We demonstrated that human ESC-MSCs showed similar morphology and cell surface markers compared with bone marrow-derived MSCs. ESC-MSCs exhibited a higher growth rate during early in vitro expansion, along with adipogenic and osteogenic differentiation potential. Treatment with ESC-MSC-conditioned medium (CM) led to statistically significant enhancement of primary hepatocyte viability and increased immunomodulatory interleukin-10 secretion from lipopolysaccharide-induced human blood mononuclear cells. Analysis of the MSCs secretome by a protein array screen showed an association between higher frequencies of secretory proteins such as vascular endothelial growth factor (VEGF) and regulation of cell proliferation, cell migration, the development process, immune system process, and apoptosis. In this thioacetamide-induced mouse model of acute liver injury, we observed that systemic infusion of VEGF led to significant survival. These data have provided the first experimental evidence of the therapeutic potential of human ESC-MSC-derived molecules. These molecules show trophic support to hepatocytes, which potentially creates new avenues for the treatment of ALF, as an inflammatory condition.


Assuntos
Células-Tronco Embrionárias Humanas/citologia , Falência Hepática Aguda/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Modelos Animais de Doenças , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Imunomodulação/efeitos dos fármacos , Falência Hepática Aguda/patologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteoma/metabolismo , Proteômica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA