Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 335
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cytometry A ; 103(9): 703-711, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37246957

RESUMEN

Very small embryonic like stem cells (VSELs) are a dormant population of stem cells that, as proposed, are deposited during embryogenesis in various tissues, including bone marrow (BM). These cells are released under steady state conditions from their tissue locations and circulate at a low level in peripheral blood (PB). Their number increases in response to stressors as well as tissue/organ damage. This increase is evident during neonatal delivery, as delivery stress prompts enrichment of umbilical cord blood (UCB) with VSELs. These cells could be purified from BM, PB, and UCB by multiparameter sorting as a population of very small CXCR4+ Lin- CD45- cells that express the CD34 or CD133 antigen. In this report, we evaluated a number of CD34+ Lin- CD45- and CD133+ Lin- CD45- UCB-derived VSELs. We also performed initial molecular characterization of both cell populations for expression of selected pluripotency markers and compared these cells at the proteomic level. We noticed that CD133+ Lin- CD45- population is more rare and express, at a higher level, mRNA for pluripotency markers Oct-4 and Nanog as well as the stromal-derived factor-1 (SDF-1) CXCR4 receptor that regulates trafficking of these cells, however both cells population did not significantly differ in the expression of proteins assigned to main biological processes.


Asunto(s)
Sangre Fetal , Proteómica , Células Madre Embrionarias , Antígenos CD34/metabolismo , Moléculas de Adhesión Celular/metabolismo
2.
Purinergic Signal ; 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37184740

RESUMEN

Hematopoiesis is regulated by several mediators such as peptide-based growth factors, cytokines, and chemokines, whose biological effects have been studied for many years. However, several other mediators have been identified recently that affect the fate of hematopoietic stem/progenitor cells (HSPC) as well as non-hematopoietic cells in the bone marrow microenvironment. These new mediators comprise members of purinergic signaling pathways and are active mediators of the soluble arm of innate immunity, the complement cascade (ComC). In this review, we will discuss the coordinated effects of these pathways in regulating the biology of HSPC. Importantly, both purinergic signaling and the ComC are activated in stress situations and interact with specific receptors expressed on HSPC. Evidence has accumulated indicating that some of the purinergic as well as ComC receptors could also be activated intracellularly by intrinsically expressed ligands. To support this recent evidence, our work indicates that the major mediator of purinergic signaling, adenosine triphosphate, and the cleavage product of the fifth component of the ComC (C5), C5a anaphylatoxin, can activate their corresponding receptors expressed on the outer mitochondrial membrane in an autocrine manner. We will also discuss recent evidence that these responses, mediated by purinergic signaling and the ComC network, are coordinated by activation of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 - reactive oxygen species - NLR family pyrin domain containing 3 (NLRP3) inflammasome (Nox2-ROS-NLRP3) axis.

3.
Mol Psychiatry ; 26(4): 1044-1059, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33328588

RESUMEN

Scientists and health professionals are exhaustively trying to contain the coronavirus disease 2019 (COVID-19) pandemic by elucidating viral invasion mechanisms, possible drugs to prevent viral infection/replication, and health cares to minimize individual exposure. Although neurological symptoms are being reported worldwide, neural acute and long-term consequences of SARS-CoV-2 are still unknown. COVID-19 complications are associated with exacerbated immunoinflammatory responses to SARS-CoV-2 invasion. In this scenario, pro-inflammatory factors are intensely released into the bloodstream, causing the so-called "cytokine storm". Both pro-inflammatory factors and viruses may cross the blood-brain barrier and enter the central nervous system, activating neuroinflammatory responses accompanied by hemorrhagic lesions and neuronal impairment, which are largely described processes in psychiatric disorders and neurodegenerative diseases. Therefore, SARS-CoV-2 infection could trigger and/or worse brain diseases. Moreover, patients with central nervous system disorders associated to neuroimmune activation (e.g. depression, Parkinson's and Alzheimer's disease) may present increased susceptibility to SARS-CoV-2 infection and/or achieve severe conditions. Elevated levels of extracellular ATP induced by SARS-CoV-2 infection may trigger hyperactivation of P2X7 receptors leading to NLRP3 inflammasome stimulation as a key mediator of neuroinvasion and consequent neuroinflammatory processes, as observed in psychiatric disorders and neurodegenerative diseases. In this context, P2X7 receptor antagonism could be a promising strategy to prevent or treat neurological complications in COVID-19 patients.


Asunto(s)
Encefalopatías/complicaciones , Encefalopatías/patología , COVID-19/complicaciones , COVID-19/patología , Neuroinmunomodulación , Receptores Purinérgicos P2X7/metabolismo , SARS-CoV-2/patogenicidad , Encefalopatías/tratamiento farmacológico , Encefalopatías/metabolismo , COVID-19/inmunología , COVID-19/metabolismo , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Pandemias , SARS-CoV-2/inmunología
4.
Curr Opin Hematol ; 28(4): 251-261, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33901136

RESUMEN

PURPOSE OF REVIEW: Hematopoiesis is co-regulated by innate immunity, which is an ancient evolutionary defense mechanism also involved in the development and regeneration of damaged tissues. This review seeks to shed more light on the workings of the Nlrp3 inflammasome, which is an intracellular innate immunity pattern recognition receptor and sensor of changes in the hematopoietic microenvironment, and focus on its role in hematopoieisis. RECENT FINDINGS: Hematopoietic stem progenitor cells (HSPCs) are exposed to several external mediators of innate immunity. Moreover, since hemato/lymphopoietic cells develop from a common stem cell, their behavior and fate are coregulated by intracellular innate immunity pathways. Therefore, the Nlrp3 inflammasome is functional both in immune cells and in HSPCs and affects hematopoiesis in either a positive or negative way, depending on its activity level. Specifically, while a physiological level of activation regulates the trafficking of HSPCs and most likely maintains their pool in the bone marrow, hyperactivation may lead to irreversible cell damage by pyroptosis and HSPC senescence and contribute to the origination of myelodysplasia and hematopoietic malignancies. SUMMARY: Modulation of the level of Nrp3 inflammasome activation will enable improvements in HSPC mobilization, homing, and engraftment strategies. It may also control pathological activation of this protein complex during HSPC senescence, graft-versus-host disease, the induction of cytokine storms, and the development of hematopoietic malignancies.


Asunto(s)
Hematopoyesis , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
5.
Purinergic Signal ; 16(2): 153-166, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32415576

RESUMEN

Bone marrow (BM) as an active hematopoietic organ is highly sensitive to changes in body microenvironments and responds to external physical stimuli from the surrounding environment. In particular, BM tissue responds to several cues related to infections, strenuous exercise, tissue/organ damage, circadian rhythms, and physical challenges such as irradiation. These multiple stimuli affect BM cells to a large degree through a coordinated response of the innate immunity network as an important guardian for maintaining homeostasis of the body. In this review, we will foc++us on the role of purinergic signaling and innate immunity in the trafficking of hematopoietic stem/progenitor cells (HSPCs) during their egression from the BM into peripheral blood (PB), as seen along pharmacological mobilization, and in the process of homing and subsequent engraftment into BM after hematopoietic transplantation. Innate immunity mediates these processes by engaging, in addition to certain peptide-based factors, other important non-peptide mediators, including bioactive phosphosphingolipids and extracellular nucleotides, as the main topic of this review. Elucidation of these mechanisms will allow development of more efficient stem cell mobilization protocols to harvest the required number of HSPCs for transplantation and to accelerate hematopoietic reconstitution in transplanted patients.


Asunto(s)
Médula Ósea/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Inmunidad Innata/inmunología , Animales , Médula Ósea/inmunología , Movimiento Celular/inmunología , Movimiento Celular/fisiología , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/inmunología , Humanos
6.
Purinergic Signal ; 16(3): 313-325, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32533388

RESUMEN

An efficient harvest of hematopoietic stem/progenitor cells (HSPCs) after pharmacological mobilization from the bone marrow (BM) into peripheral blood (PB) and subsequent proper homing and engraftment of these cells are crucial for clinical outcomes from hematopoietic transplants. Since extracellular adenosine triphosphate (eATP) plays an important role in both processes as an activator of sterile inflammation in the bone marrow microenvironment, we focused on the role of Pannexin-1 channel in the secretion of ATP to trigger both egress of HSPCs out of BM into PB as well as in reverse process that is their homing to BM niches after transplantation into myeloablated recipient. We employed a specific blocking peptide against Pannexin-1 channel and noticed decreased mobilization efficiency of HSPCs as well as other types of BM-residing stem cells including mesenchymal stroma cells (MSCs), endothelial progenitors (EPCs), and very small embryonic-like stem cells (VSELs). To explain better a role of Pannexin-1, we report that eATP activated Nlrp3 inflammasome in Gr-1+ and CD11b+ cells enriched for granulocytes and monocytes. This led to release of danger-associated molecular pattern molecules (DAMPs) and mitochondrial DNA (miDNA) that activate complement cascade (ComC) required for optimal egress of HSPCs from BM. On the other hand, Pannexin-1 channel blockage in transplant recipient mice leads to a defect in homing and engraftment of HSPCs. Based on this, Pannexin-1 channel as a source of eATP plays an important role in HSPCs trafficking.


Asunto(s)
Adenosina Trifosfato/metabolismo , Células de la Médula Ósea/metabolismo , Conexinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inflamación/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Médula Ósea/metabolismo , Inflamasomas/metabolismo , Ratones
7.
Circ Res ; 120(1): 166-178, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-28057792

RESUMEN

Evidence has accumulated that adult hematopoietic tissues and other organs contain a population of dormant stem cells (SCs) that are more primitive than other, already restricted, monopotent tissue-committed SCs (TCSCs). These observations raise several questions, such as the developmental origin of these cells, their true pluripotent or multipotent nature, which surface markers they express, how they can be efficiently isolated from adult tissues, and what role they play in the adult organism. The phenotype of these cells and expression of some genes characteristic of embryonic SCs, epiblast SCs, and primordial germ cells suggests their early-embryonic deposition in developing tissues as precursors of adult SCs. In this review, we will critically discuss all these questions and the concept that small dormant SCs related to migratory primordial germ cells, described as very small embryonic-like SCs, are deposited during embryogenesis in bone marrow and other organs as a backup population for adult tissue-committed SCs and are involved in several processes related to tissue or organ rejuvenation, aging, and cancerogenesis. The most recent results on successful ex vivo expansion of human very small embryonic-like SC in chemically defined media free from feeder-layer cells open up new and exciting possibilities for their application in regenerative medicine.


Asunto(s)
Células Madre Adultas/fisiología , Células Madre Embrionarias/fisiología , Miocitos Cardíacos/fisiología , Trasplante de Células Madre/métodos , Células Madre Adultas/trasplante , Animales , Diferenciación Celular/fisiología , Células Madre Embrionarias/trasplante , Estratos Germinativos/fisiología , Estratos Germinativos/trasplante , Humanos , Miocitos Cardíacos/trasplante , Células Madre Pluripotentes/fisiología , Células Madre Pluripotentes/trasplante
8.
Adv Exp Med Biol ; 1201: 355-388, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31898793

RESUMEN

Aging is an inevitable consequence of life, and all multicellular organisms undergo a decline in tissue and organ functions as they age. Several well-known risk factors, such as obesity, diabetes, and lack of physical activity that lead to the cardiovascular system, decline and impede the function of vital organs, ultimately limit overall life span. Over recent years, aging research has experienced an unparalleled growth, particularly with the discovery and recognition of genetic pathways and biochemical processes that control to some extent the rate of aging.In this chapter, we focus on several aspects of stem cell biology and aging, beginning with major cellular hallmarks of aging, endocrine regulation of aging and its impact on stem cell compartment, and mechanisms of increased longevity. We then discuss the role of epigenetic modifications associated with aging and provide an overview on a most recent search of antiaging modalities.


Asunto(s)
Envejecimiento/genética , Envejecimiento/metabolismo , Epigénesis Genética , Longevidad , Redes y Vías Metabólicas , Células Madre/citología , Células Madre/metabolismo , Humanos
9.
Adv Exp Med Biol ; 1201: 1-22, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31898779

RESUMEN

The field of regenerative medicine is looking for a pluripotent/multipotent stem cell able to differentiate across germ layers and be safely employed in therapy. Unfortunately, with the exception of hematopoietic stem/progenitor cells (HSPCs) for hematological applications, the current clinical results with stem cells are somewhat disappointing. The potential clinical applications of the more primitive embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have so far been discouraging, as both have exhibited several problems, including genomic instability, a risk of teratoma formation, and the possibility of rejection. Therefore, the only safe stem cells that have so far been employed in regenerative medicine are monopotent stem cells, such as the abovementioned HSPCs or mesenchymal stem cells (MSCs) isolated from postnatal tissues. However, their monopotency, and therefore limited differentiation potential, is a barrier to their broader application in the clinic. Interestingly, results have accumulated indicating that adult tissues contain rare, early-development stem cells known as very small embryonic-like stem cells (VSELs), which can differentiate into cells from more than one germ layer. This chapter addresses different sources of stem cells for potential clinical application and their advantages and problems to be solved.


Asunto(s)
Estratos Germinativos/citología , Células Madre Pluripotentes/citología , Medicina Regenerativa/tendencias , Diferenciación Celular , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
10.
Adv Exp Med Biol ; 1201: 49-77, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31898781

RESUMEN

Hematopoietic stem/progenitor cells (HSPCs) isolated from bone marrow have been successfully employed for 50 years in hematological transplantations. Currently, these cells are more frequently isolated from mobilized peripheral blood or umbilical cord blood. In this chapter, we overview several topics related to these cells including their phenotype, methods for isolation, and in vitro and in vivo assays to evaluate their proliferative potential. The successful clinical application of HSPCs is widely understood to have helped establish the rationale for the development of stem cell therapies and regenerative medicine.


Asunto(s)
Células Madre Hematopoyéticas/citología , Proliferación Celular , Separación Celular , Sangre Fetal/citología , Trasplante de Células Madre Hematopoyéticas , Humanos
11.
Adv Exp Med Biol ; 1201: 159-174, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31898786

RESUMEN

The development of regenerative medicine has provided new perspectives in many scientific fields, including psychiatry. Stem cell research is getting us closer to discovering the biological foundation of mental disorders. In this chapter, we consider the information relating to stem cells and factors involved in their trafficking in peripheral blood in some psychiatric disorders (major depressive disorder, bipolar disorder, schizophrenia, anxiety disorder, and alcohol dependence). The authors also include the implementation of current research regarding neurogenesis in adult brain and induced pluripotent stem cells in investigating concerns in etiopathogenesis of mental disorders as well as the implication of research for treatment of these disorders.


Asunto(s)
Trastornos Mentales/terapia , Psiquiatría/tendencias , Medicina Regenerativa/tendencias , Células Madre/citología , Alcoholismo , Trastornos de Ansiedad , Trastorno Bipolar , Trastorno Depresivo Mayor , Humanos , Trastornos Mentales/patología , Esquizofrenia
12.
J Cell Mol Med ; 20(1): 134-46, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26515267

RESUMEN

Evidence has accumulated that murine haematopoietic stem/progenitor cells (HSPCs) share several markers with the germline, a connection supported by recent reports that pituitary and gonadal sex hormones (SexHs) regulate development of murine HSPCs. It has also been reported that human HSPCs, like their murine counterparts, respond to certain SexHs (e.g. androgens). However, to better address the effects of SexHs, particularly pituitary SexHs, on human haematopoiesis, we tested for expression of receptors for pituitary SexHs, including follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL), as well as the receptors for gonadal SexHs, including progesterone, oestrogens, and androgen, on HSPCs purified from human umbilical cord blood (UCB) and peripheral blood (PB). We then tested the functionality of these receptors in ex vivo signal transduction studies and in vitro clonogenic assays. In parallel, we tested the effect of SexHs on human mesenchymal stromal cells (MSCs). Finally, based on our observation that at least some of the UCB-derived, CD45(-) very small embryonic-like stem cells (VSELs) become specified into CD45(+) HSPCs, we also evaluated the expression of pituitary and gonadal SexH receptors on these cells. We report for the first time that human HSPCs and VSELs, like their murine counterparts, express pituitary and gonadal SexH receptors at the mRNA and protein levels. Most importantly, SexH if added to suboptimal doses of haematopoietic cytokines and growth factors enhance clonogenic growth of human HSPCs as well as directly stimulate proliferation of MSCs.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Esteroides/metabolismo , Adhesión Celular , Proliferación Celular , Células Cultivadas , Sangre Fetal , Fibronectinas/metabolismo , Hormonas Esteroides Gonadales/fisiología , Humanos
14.
Adv Exp Med Biol ; 913: 41-49, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27796879

RESUMEN

There are several mechanisms by which cells communicate with each other. Evidence accumulates that the evolutionary oldest mechanisms of cell-cell communication involves extracellular microvesicles (ExMVs). Generally, these circular membrane fragments enriched for mRNA, miRNA, proteins, and bioactive lipids are released by exocytosis from endosomal compartment or are directly formed by budding from cell surface membranes. ExMVs from endosomal compartment called exosomes are smaller in size ~100 nM as compared to larger ones released from cell membranes that are in size up to 1 µM. In this chapter we will present an emerging link between ExMVs and recently identified novel cell-cell communication network involving a new type of cell known as telocyte. Mounting evidence accumulates that telocytes mediate several of their biological effects in several organs by releasing ExMVs enriched in mRNA, miRNA, proteins, and several biological mediators to the target cells.


Asunto(s)
Comunicación Celular/genética , Membrana Celular/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Transducción de Señal , Telocitos/metabolismo , Animales , Diferenciación Celular , Membrana Celular/ultraestructura , Vesículas Extracelulares/ultraestructura , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/ultraestructura , MicroARNs/genética , MicroARNs/metabolismo , Tamaño de la Partícula , Proteínas/genética , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Telocitos/ultraestructura
15.
J Cell Mol Med ; 19(9): 2193-201, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26033571

RESUMEN

The glycolipid glycosylphosphatidylinositol anchor (GPI-A) plays an important role in lipid raft formation, which is required for proper expression on the cell surface of two inhibitors of the complement cascade, CD55 and CD59. The absence of these markers from the surface of blood cells, including erythrocytes, makes the cells susceptible to complement lysis, as seen in patients suffering from paroxysmal nocturnal haemoglobinuria (PNH). However, the explanation for why PNH-affected hematopoietic stem/progenitor cells (HSPCs) expand over time in BM is still unclear. Here, we propose an explanation for this phenomenon and provide evidence that a defect in lipid raft formation in HSPCs leads to defective CXCR4- and VLA-4-mediated retention of these cells in BM. In support of this possibility, BM-isolated CD34(+) cells from PNH patients show a defect in the incorporation of CXCR4 and VLA-4 into membrane lipid rafts, respond weakly to SDF-1 stimulation, and show defective adhesion to fibronectin. Similar data were obtained with the GPI-A(-) Jurkat cell line. Moreover, we also report that chimeric mice transplanted with CD55(-/-)  CD59(-/-) BM cells but with proper GPI-A expression do not expand over time in transplanted hosts. On the basis of these findings, we propose that a defect in lipid raft formation in PNH-mutated HSPCs makes these cells more mobile, so that they expand and out-compete normal HSPCs from their BM niches over time.


Asunto(s)
Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/patología , Microdominios de Membrana/metabolismo , Animales , Antígenos CD/metabolismo , Toxinas Bacterianas/metabolismo , Médula Ósea/patología , Adhesión Celular/efectos de los fármacos , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Fibronectinas/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Humanos , Integrina alfa4beta1/metabolismo , Células Jurkat , Microdominios de Membrana/efectos de los fármacos , Ratones Endogámicos C57BL , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos
16.
Mol Cancer ; 14: 201, 2015 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-26597723

RESUMEN

BACKGROUND: One of the challenging problems of current radio-chemotherapy is recurrence and metastasis of cancer cells that survive initial treatment. We propose that one of the unwanted effects of radiochemotherapy is the release from damaged ("leaky") cells of nucleotides such as ATP and UTP that exert pro-metastatic functions and can directly stimulate chemotaxis of cancer cells. METHODS: To address this problem in a model of human lung cancer (LC), we employed several complementary in vitro and in vivo approaches to demonstrate the role of extracellular nucleotides (EXNs) in LC cell line metastasis and tumor progression. We measured concentrations of EXNs in several organs before and after radiochemotherapy. The purinergic receptor agonists and antagonists, inhibiting all or selected subtypes of receptors, were employed in in vitro and in vivo pro-metastatic assays. RESULTS: We found that EXNs accumulate in several organs in response to radiochemotherapy, and RT-PCR analysis revealed that most of the P1 and P2 receptor subtypes are expressed in human LC cells. EXNs were found to induce chemotaxis and adhesion of LC cells, and an autocrine loop was identified that promotes the proliferation of LC cells. Most importantly, metastasis of these cells could be inhibited in immunodeficient mice in the presence of specific small molecule inhibitors of purinergic receptors. CONCLUSIONS: Based on this result, EXNs are novel pro-metastatic factors released particularly during radiochemotherapy, and inhibition of their pro-metastatic effects via purinergic signaling could become an important part of anti-metastatic treatment.


Asunto(s)
Adenosina Trifosfato/fisiología , Factores Quimiotácticos/fisiología , Quimiotaxis , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Quimioradioterapia/efectos adversos , Líquido Extracelular/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/prevención & control , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Ratones Endogámicos C57BL , Ratones SCID , Antagonistas de Receptores Purinérgicos P1/farmacología , Antagonistas del Receptor Purinérgico P2/farmacología , Receptores Purinérgicos P1/metabolismo , Receptores Purinérgicos P2/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Stem Cells ; 32(10): 2732-43, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044556

RESUMEN

CD8(+) TCR(-) graft facilitating cells (FCs) enhance engraftment of hematopoietic stem cells (HSCs) in allogeneic and syngeneic recipients. The mechanisms by which FCs promote HSC engraftment and tolerance induction have not been fully elucidated. Here, we provide data to support a critical role for dedicator of cytokinesis 2 (DOCK2) in multiple aspects of FCs function. DOCK2(-/-) FCs exhibit compromised facilitative function in vivo as evidenced by the loss of engraftment-enhancing capability for c-Kit(+) Sca-1(+) lineage(-) (KSL) cells, and compromised ability to promote KSL cell homing and lodgment in hematopoietic niche. Deletion of DOCK2 abrogates the ability of FCs to induce differentiation of naïve CD4(+) CD25(-) T cells into FoxP3(+) regulatory T cells and interleukin-10-producing type 1 regulatory T cells in vitro. Moreover, DOCK2(-/-) FCs are unable to promote survival of KSL cells when cocultured with KSL cells. DOCK2(-/-) FCs also exhibit compromised migration to stroma-derived factor-1 in vitro and impaired homing to the bone marrow in vivo. In conclusion, our results demonstrate that DOCK2 is critical for FCs to maintain its immunomodulatory function and exert its trophic effects on KSL cells. These findings may have direct clinical relevance to promote HSC engraftment for treatment of autoimmunity, hemoglobinopathies, and to induce transplantation tolerance.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Movimiento Celular , Supervivencia Celular , Regulación hacia Abajo , Factores de Intercambio de Guanina Nucleótido , Ratones Endogámicos C57BL , Modelos Biológicos , Nicho de Células Madre , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo
18.
Stem Cells ; 32(9): 2502-15, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24806427

RESUMEN

Activation of the complement cascade (CC) with myocardial infarction (MI) acutely initiates immune cell infiltration, membrane attack complex formation on injured myocytes, and exacerbates myocardial injury. Recent studies implicate the CC in mobilization of stem/progenitor cells and tissue regeneration. Its role in chronic MI is unknown. Here, we consider complement component C3, in the chronic response to MI. C3 knockout (KO) mice were studied after permanent coronary artery ligation. C3 deficiency exacerbated myocardial dysfunction 28 days after MI compared to WT with further impaired systolic function and LV dilation despite similar infarct size 24 hours post-MI. Morphometric analysis 28 days post-MI showed C3 KO mice had more scar tissue with less viable myocardium within the infarct zone which correlated with decreased c-kit(pos) cardiac stem/progenitor cells (CPSC), decreased proliferating Ki67(pos) CSPCs and decreased formation of new BrdU(pos) /α-sarcomeric actin(pos) myocytes, and increased apoptosis compared to WT. Decreased CSPCs and increased apoptosis were evident 7 days post-MI in C3 KO hearts. The inflammatory response with MI was attenuated in the C3 KO and was accompanied by attenuated hematopoietic, pluripotent, and cardiac stem/progenitor cell mobilization into the peripheral blood 72 hours post-MI. These results are the first to demonstrate that CC, through C3, contributes to myocardial preservation and regeneration in response to chronic MI. Responses in the C3 KO infer that C3 activation in response to MI expands the resident CSPC population, increases new myocyte formation, increases and preserves myocardium, inflammatory response, and bone marrow stem/progenitor cell mobilization to preserve myocardial function.


Asunto(s)
Complemento C3/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Animales , Proliferación Celular/fisiología , Complemento C3/genética , Modelos Animales de Enfermedad , Ecocardiografía , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Regeneración/fisiología , Función Ventricular Izquierda/fisiología
19.
J Cell Mol Med ; 18(9): 1797-806, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24895014

RESUMEN

The concept that bone marrow (BM)-derived cells may participate in neural regeneration remains controversial, and the identity of the specific cell type(s) involved remains unknown. We recently reported that the adult murine BM contains a highly mobile population of Sca-1(+) Lin(-) CD45(-) cells known as very small embryonic/epiblast-like stem cells (VSELs) that express several markers of pluripotency such as Oct-4. In the BM microenvironment, these cells are kept quiescent because of epigenetic modification of certain paternally imprinted genes. However, as reported, these cells can be mobilized in mice in an experimental model of stroke and express several genes involved in neurogenesis while circulating in peripheral blood (PB). In the current work, we employed a model of toxic brain damage, which is induced by administration of kainic acid, to see not only whether VSELs can be mobilized into PB in response to this neurotoxin, but, more importantly, whether they proliferate and expand in BM tissue. We report here for the first time that brain damage leads to activation and expansion of the BM pool of quiescent VSELs, which precedes their subsequent egress into PB. Harnessing these cells in neural tissue regeneration is currently one of the challenges in regenerative medicine.


Asunto(s)
Células de la Médula Ósea/fisiología , Encefalopatías/patología , Proliferación Celular/efectos de los fármacos , Células Madre Embrionarias/fisiología , Ácido Kaínico/toxicidad , Animales , Células de la Médula Ósea/efectos de los fármacos , Encefalopatías/inducido químicamente , Movimiento Celular , Células Cultivadas , Giro Dentado/efectos de los fármacos , Giro Dentado/patología , Citometría de Flujo , Masculino , Ratones Endogámicos C57BL
20.
Blood ; 119(11): 2478-88, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22279055

RESUMEN

The mechanisms of hematopoietic progenitor cell egress and clinical mobilization are not fully understood. Herein, we report that in vivo desensitization of Sphingosine-1-phosphate (S1P) receptors by FTY720 as well as disruption of S1P gradient toward the blood, reduced steady state egress of immature progenitors and primitive Sca-1(+)/c-Kit(+)/Lin(-) (SKL) cells via inhibition of SDF-1 release. Administration of AMD3100 or G-CSF to mice with deficiencies in either S1P production or its receptor S1P(1), or pretreated with FTY720, also resulted in reduced stem and progenitor cell mobilization. Mice injected with AMD3100 or G-CSF demonstrated transient increased S1P levels in the blood mediated via mTOR signaling, as well as an elevated rate of immature c-Kit(+)/Lin(-) cells expressing surface S1P(1) in the bone marrow (BM). Importantly, we found that S1P induced SDF-1 secretion from BM stromal cells including Nestin(+) mesenchymal stem cells via reactive oxygen species (ROS) signaling. Moreover, elevated ROS production by hematopoietic progenitor cells is also regulated by S1P. Our findings reveal that the S1P/S1P(1) axis regulates progenitor cell egress and mobilization via activation of ROS signaling on both hematopoietic progenitors and BM stromal cells, and SDF-1 release. The dynamic cross-talk between S1P and SDF-1 integrates BM stromal cells and hematopoeitic progenitor cell motility.


Asunto(s)
Quimiocina CXCL12/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Bencilaminas , Médula Ósea/metabolismo , Movimiento Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Ciclamas , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Esfingosina/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA