Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 54, 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38261036

RESUMO

In embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), the expression of an RNA-binding pluripotency-relevant protein, LIN28, and the absence of its antagonist, the tumor-suppressor microRNA (miRNA) let-7, play a key role in maintaining pluripotency. Muse cells are non-tumorigenic pluripotent-like stem cells residing in the bone marrow, peripheral blood, and organ connective tissues as pluripotent surface marker SSEA-3(+). They express pluripotency genes, differentiate into triploblastic-lineage cells, and self-renew at the single cell level. Muse cells do not express LIN28 but do express let-7 at higher levels than in iPSCs. In Muse cells, we demonstrated that let-7 inhibited the PI3K-AKT pathway, leading to sustainable expression of the key pluripotency regulator KLF4 as well as its downstream genes, POU5F1, SOX2, and NANOG. Let-7 also suppressed proliferation and glycolysis by inhibiting the PI3K-AKT pathway, suggesting its involvement in non-tumorigenicity. Furthermore, the MEK/ERK pathway is not controlled by let-7 and may have a pivotal role in maintaining self-renewal and suppression of senescence. The system found in Muse cells, in which the tumor suppressor let-7, but not LIN28, tunes the expression of pluripotency genes, might be a rational cell system conferring both pluripotency-like properties and a low risk for tumorigenicity.


Assuntos
Alprostadil , Fosfatidilinositol 3-Quinases , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt , Células-Tronco Embrionárias , Expressão Gênica
2.
Cell Mol Life Sci ; 79(11): 542, 2022 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-36203068

RESUMO

Stem cells undergo cytokine-driven differentiation, but this process often takes longer than several weeks to complete. A novel mechanism for somatic stem cell differentiation via phagocytosing 'model cells' (apoptotic differentiated cells) was found to require only a short time frame. Pluripotent-like Muse cells, multipotent mesenchymal stem cells (MSCs), and neural stem cells (NSCs) phagocytosed apoptotic differentiated cells via different phagocytic receptor subsets than macrophages. The phagocytosed-differentiated cell-derived contents (e.g., transcription factors) were quickly released into the cytoplasm, translocated into the nucleus, and bound to promoter regions of the stem cell genomes. Within 24 ~ 36 h, the cells expressed lineage-specific markers corresponding to the phagocytosed-differentiated cells, both in vitro and in vivo. At 1 week, the gene expression profiles were similar to those of the authentic differentiated cells and expressed functional markers. Differentiation was limited to the inherent potential of each cell line: triploblastic-, adipogenic-/chondrogenic-, and neural-lineages in Muse cells, MSCs, and NSCs, respectively. Disruption of phagocytosis, either by phagocytic receptor inhibition via small interfering RNA or annexin V treatment, impeded differentiation in vitro and in vivo. Together, our findings uncovered a simple mechanism by which differentiation-directing factors are directly transferred to somatic stem cells by phagocytosing apoptotic differentiated cells to trigger their rapid differentiation into the target cell lineage.


Assuntos
Células-Tronco Adultas , Células-Tronco Neurais , Alprostadil , Anexina A5 , Diferenciação Celular , Citocinas , Fagocitose , RNA Interferente Pequeno , Fatores de Transcrição
3.
Surg Today ; 52(4): 603-615, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34687364

RESUMO

INTRODUCTION: We examined the effect of intravenously injected human multilineage-differentiating stress-enduring (Muse) cells, non-tumorigenic endogenous reparative stem cells already used in clinical trials, on a severe acute pancreatitis (SAP) mouse model without immunosuppressants. METHODS: Human Muse cells (1.0 × 105 cells) collected from mesenchymal stem cells (MSCs) as SSEA-3(+) were injected into a C57BL/6 mouse model via the jugular vein 6 h after SAP-induction with taurocholate. The control group received saline or the same number of SSEA-3(-)-non-Muse MSCs. RESULTS: Edematous parameters, F4/80(+) macrophage infiltration and terminal deoxynucleotidyl transferase dUTP nick-end labeling positivity was the lowest and the number of proliferating endogenous pancreatic progenitors (CK18(+)/Ki67(+) cells) the highest in the Muse group among the three groups, with statistical significance, at 72 h. An enzyme-linked immunosorbent assay and quantitative polymerase chain reaction demonstrated that in vitro production of VEGF, HGF, IGF-1, and MMP-2, which are relevant to tissue protection, anti-inflammation, and anti-fibrosis, were higher in Muse cells than in non-Muse MSCs, particularly when cells were cultured in SAP mouse serum. Consistently, the pancreas of animals in the Muse group contained higher amounts of those factors according to Western blotting at 18 h than that in the non-Muse MSCs and control groups. CONCLUSIONS: Intravenous injection of human Muse cells was suggested to be effective for attenuating edema, inflammation and apoptosis in the acute phase of SAP.


Assuntos
Imunossupressores , Pancreatite , Doença Aguda , Animais , Diferenciação Celular , Humanos , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Pancreatite/terapia
4.
Am J Transplant ; 21(6): 2025-2039, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33350582

RESUMO

Small-for-size syndrome (SFSS) has a poor prognosis due to excessive shear stress and sinusoidal microcirculatory disturbances in the acute phase after living-donor liver transplantation (LDLT). Multilineage-differentiating stress enduring (Muse) cells are reparative stem cells found in various tissues and currently under clinical trials. These cells selectively home to damaged sites via the sphingosine-1-phosphate (S1P)-S1P receptor 2 system and repair damaged tissue by pleiotropic effects, including tissue protection and damaged/apoptotic cell replacement by differentiating into tissue-constituent cells. The effects of intravenously administered human bone marrow-Muse cells and -mesenchymal stem cells (MSCs) (4 × 105 ) on liver sinusoidal endothelial cells (LSECs) were examined in a rat SFSS model without immunosuppression. Compared with MSCs, Muse cells intensively homed to the grafted liver, distributed to the sinusoids and vessels, and delivered improved blood chemistry and Ki-67(+) proliferative hepatocytes and -LSECs within 3 days. Tissue clearing and three-dimensional imaging by multiphoton laser confocal microscopy revealed maintenance of the sinusoid continuity, organization, and surface area, as well as decreased sinusoid interruption in the Muse group. Small-interfering RNA-induced knockdown of hepatocyte growth factor and vascular endothelial growth factor-A impaired the protective effect of Muse cells on LSECs. Intravenous injection of Muse cells might be a feasible approach for LDLT with less recipient burden.


Assuntos
Transplante de Fígado , Alprostadil , Animais , Capilares , Diferenciação Celular , Células Endoteliais , Humanos , Infusões Intravenosas , Fígado , Doadores Vivos , Microcirculação , Ratos , Fator A de Crescimento do Endotélio Vascular
5.
Mol Ther ; 28(1): 100-118, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31607541

RESUMO

Shiga toxin-producing Escherichia coli (STEC) causes hemorrhagic colitis, hemolytic uremic syndrome, and acute encephalopathies that may lead to sudden death or severe neurologic sequelae. Current treatments, including immunoglobulin G (IgG) immunoadsorption, plasma exchange, steroid pulse therapy, and the monoclonal antibody eculizumab, have limited effects against the severe neurologic sequelae. Multilineage-differentiating stress-enduring (Muse) cells are endogenous reparative non-tumorigenic stem cells that naturally reside in the body and are currently under clinical trials for regenerative medicine. When administered intravenously, Musecells accumulate to the damaged tissue, where they exert anti-inflammatory, anti-apoptotic, anti-fibrotic, and immunomodulatory effects, and replace damaged cells by differentiating into tissue-constituent cells. Here, severely immunocompromised non-obese diabetic/severe combined immunodeficiency (NOD-SCID) mice orally inoculated with 9 × 109 colony-forming units of STEC O111 and treated 48 h later with intravenous injection of 5 × 104 Muse cells exhibited 100% survival and no severe after-effects of infection. Suppression of granulocyte-colony-stimulating factor (G-CSF) by RNAi abolished the beneficial effects of Muse cells, leading to a 40% death and significant body weight loss, suggesting the involvement of G-CSF in the beneficial effects of Muse cells in STEC-infected mice. Thus, intravenous administration of Muse cells could be a candidate therapeutic approach for preventing fatal encephalopathy after STEC infection.


Assuntos
Encefalopatias/microbiologia , Encefalopatias/terapia , Transplante de Células/métodos , Infecções por Escherichia coli/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Toxina Shiga II/metabolismo , Escherichia coli Shiga Toxigênica/metabolismo , Adulto , Idoso de 80 Anos ou mais , Animais , Encéfalo/patologia , Encefalopatias/epidemiologia , Encefalopatias/metabolismo , Modelos Animais de Doenças , Surtos de Doenças , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Feminino , Humanos , Injeções Intravenosas , Japão/epidemiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Endogâmicos NOD , Camundongos SCID , Resultado do Tratamento
6.
Surg Today ; 51(4): 634-650, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32915286

RESUMO

INTRODUCTION: Multilineage-differentiating stress-enduring (Muse) cells are non-tumorigenic endogenous pluripotent-like cells residing in the bone marrow that exert a tissue reparative effect by replacing damaged/apoptotic cells through spontaneous differentiation into tissue-constituent cells. Post-hepatectomy liver failure (PHLF) is a potentially fatal complication. The main purpose of this study was to evaluate the safety and efficiency of allogeneic Muse cell administration via the portal vein in a swine model of PHLF. METHODS: Swine Muse cells, collected from swine bone marrow-mesenchymal stem cells (MSCs) as SSEA-3(+) cells, were examined for their characteristics. Then, 1 × 107 allogeneic-Muse cells and allogeneic-MSCs and vehicle were injected via the portal vein in a 70% hepatectomy swine model. RESULTS: Swine Muse cells exhibited characteristics comparable to previously reported human Muse cells. Compared to the MSC and vehicle groups, the Muse group showed specific homing of the administered cells into the liver, resulting in improvements in the control of hyperbilirubinemia (P = 0.04), prothrombin international normalized ratio (P = 0.05), and suppression of focal necrosis (P = 0.04). Integrated Muse cells differentiated spontaneously into hepatocyte marker-positive cells. CONCLUSIONS: Allogeneic Muse cell administration may provide a reparative effect and functional recovery in a 70% hepatectomy swine model and thus may contribute to the treatment of PHLF.


Assuntos
Hepatectomia/efeitos adversos , Falência Hepática/etiologia , Falência Hepática/terapia , Transplante de Células-Tronco Mesenquimais/métodos , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/terapia , Animais , Modelos Animais de Doenças , Veia Porta , Recuperação de Função Fisiológica , Segurança , Suínos , Transplante Homólogo , Resultado do Tratamento
7.
Stroke ; 51(2): 601-611, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31826733

RESUMO

Background and Purpose- Multilineage-differentiating stress-enduring cells are endogenous nontumorigenic reparative pluripotent-like stem cells found in bone marrow, peripheral blood, and connective tissues. Topically administered human multilineage-differentiating stress-enduring cells into rat/mouse stroke models differentiated into neural cells and promoted clinically relevant functional recovery. However, critical questions on the appropriate timing and dose, and safety of the less invasive intravenous administration of clinical-grade multilineage-differentiating stress-enduring cell-based product CL2020 remain unanswered. Methods- Using an immunodeficient mouse lacunar model, CL2020 was administered via the cervical vein in different doses (high dose=5×104 cells/body; medium dose=1×104 cells/body; low dose=5×103 cells/body) at subacute phase (≈9 days after onset) and chronic phase (≈30 days). Cylinder test, depletion of human cells by diphtheria toxin administration, immunohistochemistry, and human specific-genome detection were performed. Results- Tumorigenesis and adverse effects were not detected for up to 22 weeks. The high-dose group displayed significant functional recovery compared with the vehicle group in cylinder test in subacute-phase-treated and chronic-phase-treated animals after 6 weeks and 8 weeks post-injection, respectively. In the high-dose group of subacute-phase-treated animals, robust and stable recovery in cylinder test persisted up to 22 weeks compared with the vehicle group. In both groups, intraperitoneal injection of diphtheria toxin abrogated the functional recovery. Anti-human mitochondria revealed CL2020 distributed mainly in the peri-infarct area at 1, 10, and 22 weeks and expressed NeuN (neuronal nuclei)- and MAP-2 (microtubule-associated protein-2)-immunoreactivity. Conclusions- Intravenously administered CL2020 was safe, migrated to the peri-infarct area, and afforded functional recovery in experimental stroke.


Assuntos
Transplante de Células-Tronco , Acidente Vascular Cerebral Lacunar , Acidente Vascular Cerebral/cirurgia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Movimento Celular/fisiologia , Modelos Animais de Doenças , Camundongos Transgênicos , Recuperação de Função Fisiológica/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral Lacunar/fisiopatologia , Acidente Vascular Cerebral Lacunar/terapia
8.
Circ Res ; 122(8): 1069-1083, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29475983

RESUMO

RATIONALE: Multilineage-differentiating stress enduring (Muse) cells, pluripotent marker stage-specific embryonic antigen-3+ cells, are nontumorigenic endogenous pluripotent-like stem cells obtainable from various tissues including the bone marrow. Their therapeutic efficiency has not been validated in acute myocardial infarction. OBJECTIVE: The main objective of this study is to clarify the efficiency of intravenously infused rabbit autograft, allograft, and xenograft (human) bone marrow-Muse cells in a rabbit acute myocardial infarction model and their mechanisms of tissue repair. METHODS AND RESULTS: In vivo dynamics of Nano-lantern-labeled Muse cells showed preferential homing of the cells to the postinfarct heart at 3 days and 2 weeks, with ≈14.5% of injected GFP (green fluorescent protein)-Muse cells estimated to be engrafted into the heart at 3 days. The migration and homing of the Muse cells was confirmed pharmacologically (S1PR2 [sphingosine monophosphate receptor 2]-specific antagonist JTE-013 coinjection) and genetically (S1PR2-siRNA [small interfering ribonucleic acid]-introduced Muse cells) to be mediated through the S1P (sphingosine monophosphate)-S1PR2 axis. They spontaneously differentiated into cells positive for cardiac markers, such as cardiac troponin-I, sarcomeric α-actinin, and connexin-43, and vascular markers. GCaMP3 (GFP-based Ca calmodulin probe)-labeled Muse cells that engrafted into the ischemic region exhibited increased GCaMP3 fluorescence during systole and decreased fluorescence during diastole. Infarct size was reduced by ≈52%, and the ejection fraction was increased by ≈38% compared with vehicle injection at 2 months, ≈2.5 and ≈2.1 times higher, respectively, than that induced by mesenchymal stem cells. These effects were partially attenuated by the administration of GATA4-gene-silenced Muse cells. Muse cell allografts and xenografts efficiently engrafted and recovered functions, and allografts remained in the tissue and sustained functional recovery for up to 6 months without immunosuppression. CONCLUSIONS: Muse cells may provide reparative effects and robust functional recovery and may, thus, provide a novel strategy for the treatment of acute myocardial infarction.


Assuntos
Lisofosfolipídeos/fisiologia , Infarto do Miocárdio/cirurgia , Células-Tronco Pluripotentes/transplante , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Aloenxertos , Animais , Autoenxertos , Diferenciação Celular , Movimento Celular/fisiologia , Fator de Transcrição GATA4/antagonistas & inibidores , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/fisiologia , Sobrevivência de Enxerto , Proteínas de Fluorescência Verde/análise , Xenoenxertos , Humanos , Luciferases/análise , Proteínas Luminescentes/análise , Masculino , Infarto do Miocárdio/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Coelhos , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes de Fusão/análise , Especificidade da Espécie , Esfingosina/fisiologia , Receptores de Esfingosina-1-Fosfato
9.
Glia ; 67(5): 950-966, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30637802

RESUMO

Direct conversion is considered a promising approach to obtain tissue-specific cells for cell therapies; however, this strategy depends on exogenous gene expression that may cause undesired adverse effects such as tumorigenesis. By optimizing the Schwann cell induction system, which was originally developed for trans-differentiation of bone marrow mesenchymal stem cells into Schwann cells, we established a system to directly convert adult human skin fibroblasts into cells comparable to authentic human Schwann cells without gene introduction. Serial treatments with beta-mercaptoethanol, retinoic acid, and finally a cocktail of basic fibroblast growth factor, forskolin, platelet-derived growth factor-AA, and heregulin-ß1 (EGF domain) converted fibroblasts into cells expressing authentic Schwann cell markers at an efficiency of approximately 75%. Genome-wide gene expression analysis suggested the conversion of fibroblasts into the Schwann cell-lineage. Transplantation of induced Schwann cells into severed peripheral nerve of rats facilitated axonal regeneration and robust functional recovery in sciatic function index comparable to those of authentic human Schwann cells. The contributions of induced Schwann cells to myelination of regenerated axons and re-formation of neuromuscular junctions were also demonstrated. Our data clearly demonstrated that cells comparable to functional Schwann cells feasible for the treatment of neural disease can be induced from adult human skin fibroblasts without gene introduction. This direct conversion system will be beneficial for clinical applications to peripheral and central nervous system injuries and demyelinating diseases.


Assuntos
Diferenciação Celular/fisiologia , Fibroblastos/fisiologia , Traumatismos dos Nervos Periféricos/cirurgia , Recuperação de Função Fisiológica/fisiologia , Células de Schwann/fisiologia , Células de Schwann/transplante , Animais , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Humanos , Locomoção/fisiologia , Masculino , Microscopia Eletrônica , Proteína P0 da Mielina/metabolismo , Traumatismos dos Nervos Periféricos/fisiopatologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fatores de Transcrição SOXE/metabolismo , Células de Schwann/ultraestrutura , Soro/fisiologia , Pele/citologia , Fatores de Tempo , Tretinoína/farmacologia
10.
Dev Growth Differ ; 60(6): 326-340, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29984494

RESUMO

Proliferation of ependymal cells of the adult spinal cord (SCEp cells) in the intact condition has been considered as a quite rare event. To visualize proliferating/proliferated SCEp cells, we used the intensive 5-bromo-2'-deoxyuridine (BrdU) administration method to find that about two cells in the ependymal layer incorporated BrdU in a 10-µm-thick section. Because these two cells were not considered to undergo further proliferation, we analyzed the positioning and motility of two neighboring BrdU-incorporated proliferated cells and elucidated the tendency of the movement of SCEp cells to the outer side inside the ependymal layer. Even if it was rare, one of the proliferated cells in the ependymal layer differentiated into astrocytes. Gene introduction of Notch intracellular domain (NICD), a constitutively active form of Notch1, into SCEp cells demonstrated both increase in proliferation and induction of differentiation into astrocytes. Overexpression of Sox2 promoted proliferation in SCEp cells. The reaction of gene introduction of NICD and Sox2 indicates the similarity of intracellular signaling between SCEp cells and neural stem cells. Also, considering the fact that SCEp cells express these two factors in the intact condition, Notch and Sox2 are important for the cell fate control of SCEp cells in the intact condition.


Assuntos
Astrócitos/metabolismo , Diferenciação Celular , Proliferação de Células , Epêndima/metabolismo , Células-Tronco Neurais/metabolismo , Transdução de Sinais/fisiologia , Animais , Antígenos de Diferenciação/biossíntese , Antígenos de Diferenciação/fisiologia , Astrócitos/citologia , Epêndima/citologia , Regulação da Expressão Gênica , Masculino , Células-Tronco Neurais/citologia , Ratos , Ratos Wistar , Fatores de Transcrição SOXB1
11.
Adv Exp Med Biol ; 1103: C1, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31236853

RESUMO

The below listed corrections have been carried out in the following pages of the current version.

12.
Adv Exp Med Biol ; 1103: 13-41, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30484222

RESUMO

Multilineage-differentiating stress-enduring (Muse) cells exhibit the core characteristics of pluripotent stem cells, namely, the expression of pluripotency markers and the capacity for trilineage differentiation both in vitro and in vivo and self-renewability. In addition, Muse cells have unique characteristics not observed in other pluripotent stem cells such as embryonic stem cells, control of pluripotency by environmental switch of adherent suspension, symmetric and asymmetric cell division, expression of factors relevant to stress tolerance, and distinctive tissue distribution. Pluripotent stem cells were recently classified into two discrete states, naïve and primed. These two states have multiple functional differences, including their proliferation rate, molecular properties, and growth factor dependency. The properties exhibited by Muse cells are similar to those of primed pluripotent stem cells while with some uniqueness. In this chapter, we provide a comprehensive description of the basic characteristics of Muse cells.


Assuntos
Linhagem da Célula , Células-Tronco Pluripotentes/citologia , Estresse Fisiológico , Diferenciação Celular , Proliferação de Células , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia
13.
Adv Exp Med Biol ; 1103: 43-68, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30484223

RESUMO

The dynamics and actions of Muse cells at a time of physical crisis are unique and highly remarkable compared with other stem cell types. When the living body is in a steady state, low levels of Muse cells are mobilized to the peripheral blood, possibly from the bone marrow, and supplied to the connective tissue of nearly every organ. Under conditions of serious tissue damage, such as acute myocardial infarction and stroke, Muse cells are highly mobilized to the peripheral blood, drastically increasing their numbers in the peripheral blood within 24 h after the onset of tissue injury. The alerting signal, sphingosine-1-phosphate, attracts Muse cells to the damaged site mainly via the sphingosine-1-phosphate receptor 2, enabling them to preferentially home to site of injury. After homing, Muse cells spontaneously differentiate into tissue-compatible cells and replenish new functional cells for tissue repair. Because Muse cells have pleiotropic effects, including paracrine, anti-inflammatory, anti-fibrotic, and anti-apoptotic effects, these cells synergistically deliver long-lasting functional and structural recovery. This chapter describes how Muse cells exert their reparative effects in vivo.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Regeneração , Ferimentos e Lesões/fisiopatologia , Movimento Celular , Humanos , Lisofosfolipídeos , Receptores de Lisoesfingolipídeo , Esfingosina/análogos & derivados
14.
Adv Exp Med Biol ; 1103: 69-101, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30484224

RESUMO

This chapter provides the detailed method for isolation of Muse cells and evaluation of their pluripotency. The basic population of Muse cells is cultured mesenchymal stem cells such as bone marrow-mesenchymal stem cells, fibroblasts, and adipose-derived stem cells. The detailed method for handling mesenchymal stem cells is also provided in this protocol.


Assuntos
Separação Celular/métodos , Células-Tronco Pluripotentes/citologia , Adipócitos/citologia , Células da Medula Óssea/citologia , Células Cultivadas , Fibroblastos/citologia , Humanos , Células-Tronco Mesenquimais/citologia
15.
J Am Soc Nephrol ; 28(10): 2946-2960, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28674043

RESUMO

Multilineage-differentiating stress-enduring (Muse) cells are nontumorigenic endogenous pluripotent-like stem cells that can be collected from various organs. Intravenously administered Muse cells have been shown to spontaneously migrate to damaged tissue and replenish lost cells, but the effect in FSGS is unknown. We systemically administered human bone marrow-derived Muse cells without concurrent administration of immunosuppressants to severe combined immune-deficient (SCID) and BALB/c mouse models with adriamycin-induced FSGS (FSGS-SCID and FSGS-BALB/c, respectively). In FSGS-SCID mice, human Muse cells preferentially integrated into the damaged glomeruli and spontaneously differentiated into cells expressing markers of podocytes (podocin; 31%), mesangial cells (megsin; 13%), and endothelial cells (CD31; 41%) without fusing to the host cells; attenuated glomerular sclerosis and interstitial fibrosis; and induced the recovery of creatinine clearance at 7 weeks. Human Muse cells induced similar effects in FSGS-BALB/c mice at 5 weeks, despite xenotransplant without concurrent immunosuppressant administration, and led to improvement in urine protein, creatinine clearance, and plasma creatinine levels more impressive than that in the FSGS-SCID mice at 5 weeks. However, functional recovery in FSGS-BALB/c mice was impaired at 7 weeks due to immunorejection, suggesting the importance of Muse cell survival as glomerular cells in the FSGS kidney for tissue repair and functional recovery. In conclusion, Muse cells are unique reparative stem cells that preferentially home to damaged glomeruli and spontaneously differentiate into glomerular cells after systemic administration. Introduction of genes to induce differentiation is not required before Muse cell administration; thus, Muse cells may be a feasible therapeutic strategy in FSGS.


Assuntos
Glomerulosclerose Segmentar e Focal/terapia , Transplante de Células-Tronco , Animais , Diferenciação Celular , Movimento Celular , Doxorrubicina , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Humanos , Testes de Função Renal , Camundongos Endogâmicos BALB C , Camundongos SCID , Regeneração
16.
Stroke ; 48(2): 428-435, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27999136

RESUMO

BACKGROUND AND PURPOSE: Multilineage-differentiating stress-enduring (muse) cells are endogenous nontumorigenic stem cells with pluripotency harvestable as pluripotent marker SSEA-3+ cells from the bone marrow from cultured bone marrow-mesenchymal stem cells. After transplantation into neurological disease models, muse cells exert repair effects, but the exact mechanism remains inconclusive. METHODS: We conducted mechanism-based experiments by transplanting serum/xeno-free cultured-human bone marrow-muse cells into the perilesion brain at 2 weeks after lacunar infarction in immunodeficient mice. RESULTS: Approximately 28% of initially transplanted muse cells remained in the host brain at 8 weeks, spontaneously differentiated into cells expressing NeuN (≈62%), MAP2 (≈30%), and GST-pi (≈12%). Dextran tracing revealed connections between host neurons and muse cells at the lesioned motor cortex and the anterior horn. Muse cells extended neurites through the ipsilateral pyramidal tract, crossed to contralateral side, and reached to the pyramidal tract in the dorsal funiculus of spinal cord. Muse-transplanted stroke mice displayed significant recovery in cylinder tests, which was reverted by the human-selective diphtheria toxin. At 10 months post-transplantation, human-specific Alu sequence was detected only in the brain but not in other organs, with no evidence of tumor formation. CONCLUSIONS: Transplantation at the delayed subacute phase showed muse cells differentiated into neural cells, facilitated neural reconstruction, improved functions, and displayed solid safety outcomes over prolonged graft maturation period, indicating their therapeutic potential for lacunar stroke.


Assuntos
Encéfalo/fisiologia , Modelos Animais de Doenças , Transplante de Células-Tronco Mesenquimais/métodos , Rede Nervosa/fisiologia , Acidente Vascular Cerebral Lacunar/terapia , Animais , Encéfalo/citologia , Encéfalo/patologia , Linhagem da Célula , Humanos , Masculino , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Acidente Vascular Cerebral Lacunar/patologia
17.
Stem Cells ; 34(1): 160-73, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26388204

RESUMO

OBJECTIVE: Muse cells reside as pre-existing pluripotent-like stem cells within the fibroblasts, are nontumorigenic, exhibit differentiation capacity into triploblastic-lineage cells, and replenish lost cells when transplanted in injury models. Cell fate and function of human skin fibroblast-derived Muse cells were evaluated in a rat stroke model. METHODS: Muse cells (30,000), collected by pluripotent surface marker stage-specific embryonic antigen-3, were injected stereotaxically into three deposits within the rat ischemic cortex at 2 days after transient middle cerebral artery occlusion, and the cells' biological effects were examined for more than 84 days. RESULTS: Muse cells spontaneously and promptly committed to neural/neuronal-lineage cells when cocultured with stroke brain slices. Muse-transplanted stroke rats exhibited significant improvements in neurological and motor functions compared to control groups at chronic days 70 and 84, without a reduction in the infarct size. Muse cells survived in the host brain for up to 84 days and differentiated into NeuN (∼ 65%), MAP-2 (∼ 32%), calbindin (∼ 28%), and GST-π (∼ 25%)-positive cells in the cortex, but glial fibrillary acidic protein-positive cells were rare. Tumor formation was not observed. Muse cells integrated into the sensory-motor cortex, extended their neurites into cervical spinal cord, and displayed normalized hind limb somatosensory evoked potentials. INTERPRETATION: Muse cells are unique from other stem cells in that they differentiate with high ratio into neuronal cells after integration with host brain microenvironment, possibly reconstructing the neuronal circuit to mitigate stroke symptoms. Human fibroblast-derived Muse cells pose as a novel source of transplantable stem cells, circumventing the need for gene manipulations, especially when contemplating autologous cell therapy for stroke.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Fibroblastos/transplante , Neurônios/citologia , Acidente Vascular Cerebral/terapia , Adulto , Animais , Comportamento Animal , Encéfalo/patologia , Linhagem da Célula , Sobrevivência Celular , Microambiente Celular , Fenômenos Eletrofisiológicos , Humanos , Camundongos SCID , Córtex Motor/patologia , Ratos , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/fisiopatologia
18.
Exp Brain Res ; 235(2): 565-572, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27817105

RESUMO

A novel type of non-tumorigenic pluripotent stem cell, the Muse cell (multi-lineage, differentiating stress enduring cell), resides in the connective tissue and in cultured mesenchymal stem cells (MSCs) and is reported to differentiate into multiple cell types according to the microenvironment to repair tissue damage. We examined the efficiency of Muse cells in a mouse intracerebral hemorrhage (ICH) model. Seventy µl of cardiac blood was stereotactically injected into the left putamen of immunodeficient mice. Five days later, 2 × 105 of human bone marrow MSC-derived Muse cells (n = 6) or cells other than Muse cells in MSCs (non-Muse, n = 6) or the same volume of PBS (n = 11) was injected into the ICH cavity. Water maze and motor function tests were implemented for 68 days, and immunohistochemistry for NeuN, MAP2 and GFAP was done. The Muse group showed impressive recovery: Recovery was seen in the water maze after day 19, and motor functions after 5 days was compared with the other two groups, with a significant statistical difference (p < 0.05). The survival rate of the engrafted cells in the Muse group was significantly higher than in the non-Muse group (p < 0.05) at day 69, and those cells showed positivity for NeuN (~57%) and MAP-2 (~41.6%). Muse cells could remain in the ICH brain, differentiate into neural-lineage cells and restore functions without inducing them into neuronal cells by gene introduction and cytokine treatment prior to transplantation. A simple collection of Muse cells and their supply to the brain in naïve state facilitates regenerative therapy in ICH.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Hemorragia Cerebral/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Degeneração Neural/cirurgia , Análise de Variância , Animais , Linhagem Celular , Hemorragia Cerebral/complicações , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos SCID , Degeneração Neural/etiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Exame Neurológico , Medicina Regenerativa/métodos , Fatores de Tempo
19.
Cells Tissues Organs ; 200(1): 31-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25765009

RESUMO

Schwann cells form myelin, sustain axons and provide the microenvironment for nerve fibers, thereby playing a key role in the peripheral nervous system (PNS). Schwann cells also provide support for the damaged PNS by producing factors that strongly promote axonal regrowth and contribute to remyelination, which is crucial for the recovery of neural function. These advantages are not confined to the PNS and also apply to the central nervous system. Many diseases, including peripheral nerve injury, neuropathy, multiple sclerosis and spinal cord injury, are targets for Schwann cell therapy. The collection of Schwann cells, however, causes new damage to other peripheral nerve segments. Furthermore, the doubling time of Schwann cells is not very fast, and thus adequate amounts of Schwann cells for clinical use cannot be collected within a reasonable amount of time. Mesenchymal stem cells, which are highly proliferative, are easily accessible from various types of mesenchymal tissues, such as the bone marrow, umbilical cord and fat tissue. Because these cells have the ability to cross oligolineage boundaries between mesodermal to ectodermal lineages, they are capable of differentiating into Schwann cells with step-by-step cytokine stimulation. In this review, we summarize the properties of mesenchymal stem cell-derived Schwann cells, which are comparable to authentic Schwann cells, and discuss future perspectives.


Assuntos
Células-Tronco Mesenquimais/citologia , Regeneração Nervosa , Nervos Periféricos/fisiopatologia , Células de Schwann/citologia , Animais , Axônios/metabolismo , Modelos Animais de Doenças , Humanos
20.
Pathol Int ; 64(1): 1-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24471964

RESUMO

Mesenchymal stem cells (MSCs) have been presumed to include a subpopulation of pluripotent-like cells as they differentiate not only into the same mesodermal-lineage cells but also into ectodermal- and endodermal-lineage cells and exert tissue regenerative effects in a wide variety of tissues. A novel type of pluripotent stem cell, Multilineage-differentiating stress enduring (Muse) cells, was recently discovered in mesenchymal tissues such as the bone marrow, adipose tissue, dermis and connective tissue of organs, as well as in cultured fibroblasts and bone marrow-MSCs. Muse cells are able to differentiate into all three germ layers from a single cell and to self-renew, and yet exhibit non-tumorigenic and low telomerase activities. They can migrate to and target damaged sites in vivo, spontaneously differentiate into cells compatible with the targeted tissue, and contribute to tissue repair. Thus, Muse cells may account for the wide variety of differentiation abilities and tissue repair effects that have been observed in MSCs. Muse cells are unique in that they are pluripotent stem cells that belong in the living body, and are thus assumed to play an important role in 'regenerative homeostasis' in vivo.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Linhagem da Célula/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Pluripotentes/citologia , Animais , Carcinogênese/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA