Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
1.
Purinergic Signal ; 16(2): 153-166, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32415576

RESUMO

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.


Assuntos
Medula Óssea/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Imunidade Inata/imunologia , Animais , Medula Óssea/imunologia , Movimento Celular/imunologia , Movimento Celular/fisiologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/imunologia , Humanos
2.
Circ Res ; 120(1): 166-178, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-28057792

RESUMO

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.


Assuntos
Células-Tronco Adultas/fisiologia , Células-Tronco Embrionárias/fisiologia , Miócitos Cardíacos/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco Adultas/transplante , Animais , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/transplante , Camadas Germinativas/fisiologia , Camadas Germinativas/transplante , Humanos , Miócitos Cardíacos/transplante , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante
3.
Adv Exp Med Biol ; 1201: 49-77, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31898781

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/citologia , Proliferação de Células , Separação Celular , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Humanos
4.
J Cell Mol Med ; 20(1): 134-46, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26515267

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Esteroides/metabolismo , Adesão Celular , Proliferação de Células , Células Cultivadas , Sangue Fetal , Fibronectinas/metabolismo , Hormônios Esteroides Gonadais/fisiologia , Humanos
6.
J Cell Mol Med ; 19(9): 2193-201, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26033571

RESUMO

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.


Assuntos
Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/patologia , Microdomínios da Membrana/metabolismo , Animais , Antígenos CD/metabolismo , Toxinas Bacterianas/metabolismo , Medula Óssea/patologia , Adesão Celular/efeitos dos fármacos , Quimiocina CXCL12/farmacologia , Quimiotaxia/efeitos dos fármacos , Fibronectinas/metabolismo , Glicosilfosfatidilinositóis/metabolismo , Humanos , Integrina alfa4beta1/metabolismo , Células Jurkat , Microdomínios da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
J Cell Mol Med ; 18(9): 1797-806, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24895014

RESUMO

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.


Assuntos
Células da Medula Óssea/fisiologia , Encefalopatias/patologia , Proliferação de Células/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Ácido Caínico/toxicidade , Animais , Células da Medula Óssea/efeitos dos fármacos , Encefalopatias/induzido quimicamente , Movimento Celular , Células Cultivadas , Giro Denteado/efeitos dos fármacos , Giro Denteado/patologia , Citometria de Fluxo , Masculino , Camundongos Endogâmicos C57BL
8.
Stem Cells ; 31(3): 500-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23193025

RESUMO

Ceramide-1-phosphate (C1P) is a bioactive lipid that, in contrast to ceramide, is an antiapoptotic molecule released from cells that are damaged and "leaky." As reported recently, C1P promotes migration of hematopoietic cells. In this article, we tested the hypothesis that C1P released upon tissue damage may play an underappreciated role in chemoattraction of various types of stem cells and endothelial cells involved in tissue/organ regeneration. We show for the first time that C1P is upregulated in damaged tissues and chemoattracts bone marrow (BM)-derived multipotent stromal cells, endothelial progenitor cells, and very small embryonic-like stem cells. Furthermore, compared to other bioactive lipids, C1P more potently chemoattracted human umbilical vein endothelial cells and stimulated tube formation by these cells. C1P also promoted in vivo vascularization of Matrigel implants and stimulated secretion of stromal cell-derived factor-1 from BM-derived fibroblasts. Thus, our data demonstrate, for the first time, that C1P is a potent bioactive lipid released from damaged cells that potentially plays an important and novel role in recruitment of stem/progenitor cells to damaged organs and may promote their vascularization.


Assuntos
Movimento Celular/fisiologia , Ceramidas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Processos de Crescimento Celular/fisiologia , Ceramidas/biossíntese , Fatores Quimiotáticos/biossíntese , Fatores Quimiotáticos/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Medicina Regenerativa/métodos , Células-Tronco/metabolismo , Regulação para Cima
9.
Stem Cell Rev Rep ; 20(1): 237-246, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37812364

RESUMO

Hematopoietic stem progenitor cells (HSPCs) follow the diurnal circulation rhythm in peripheral blood (PB) with nadir during late night and peak at early morning hours. The level of these cells in PB correlates with activation of innate immunity pathways, including complement cascade (ComC) that drives activation of Nlrp3 inflammasome. To support this, mice both in defective ComC activation as well as Nlrp3 inflammasome do not show typical changes in the diurnal level of circulating HSPCs. Migration of HSPCs is also impaired at the intracellular level by the anti-inflammatory enzyme heme oxygenase-1 (HO-1) which is an inhibitor of Nlrp3 inflammasome. It is also well known that circadian rhythm mediates PB level of melatonin released from the pineal gland. Since trafficking of HSPCs is driven by innate immunity-induced sterile inflammation and melatonin has an anti-inflammatory effect, we hypothesized that melatonin could negatively impact the release of HSPCs from BM into PB by inhibiting Nlrp3 inflammasome activation. We provide an evidence that melatonin being a ''sleep regulating pineal hormone'' directly inhibits migration of HSPCs both in vitro migration assays and in vivo during pharmacological mobilization. This correlated with inhibition of cholesterol synthesis required for a proper membrane lipid raft (MLRs) formation and an increase in expression of HO-1-an inhibitor of Nlrp3 inflammasome. Since melatonin is a commonly used drug, this should be considered while preparing a patient for the procedure of HSPCs mobilization. More importantly, our studies shed more mechanistic light on a role of melatonin in the diurnal circulation of HSPCs.


Assuntos
Melatonina , Glândula Pineal , Humanos , Animais , Camundongos , Inflamassomos/metabolismo , Melatonina/farmacologia , Melatonina/metabolismo , Glândula Pineal/metabolismo , Heme Oxigenase-1/metabolismo , Células-Tronco Hematopoéticas , Anti-Inflamatórios , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
10.
Adv Exp Med Biol ; 777: 125-41, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23161080

RESUMO

CD133 antigen (prominin-1) is a useful cell surface marker of very small embryonic-like stem cells (VSELs). Antibodies against it, conjugated to paramagnetic beads or fluorochromes, are thus powerful biological tools for their isolation from human umbilical cord blood, mobilized peripheral blood, and bone marrow. VSELs are described with the following characteristics: (1) are slightly smaller than red blood cells; (2) display a distinct morphology, typified by a high nuclear/cytoplasmic ratio and an unorganized euchromatin; (3) become mobilized during stress situations into peripheral blood; (4) are enriched in the CD133(+)Lin(-)CD45(-) cell fraction in humans; and (5) express markers of pluripotent stem cells (e.g., Oct-4, Nanog, and stage-specific embryonic antigen-4). The most recent in vivo data from our and other laboratories demonstrated that human VSELs exhibit some characteristics of long-term repopulating hematopoietic stem cells and are at the top of the hierarchy in the mesenchymal lineage. However, still more labor is needed to characterize better at a molecular level these rare cells.


Assuntos
Camadas Germinativas , Células-Tronco Pluripotentes , Células-Tronco Embrionárias/citologia , Sangue Fetal , Células-Tronco Hematopoéticas/citologia , Humanos , Fenótipo , Células-Tronco Pluripotentes/citologia
11.
Adv Exp Med Biol ; 946: 37-54, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21948361

RESUMO

Hematopoietic stem and progenitor cells (HSPCs) circulate under steady-state conditions at detectable levels in peripheral blood (PB). The phenomenon of enforced release of HSPCs from BM into PB is called mobilization and may be envisioned as a danger-sensing response mechanism triggered by hypoxia or mechanical- or infection-induced tissue damage and is a part of stress response. It is unquestionable that the a-chemokine stromal derived factor-1 (SDF-1)-CXCR4 axis plays crucial role in retention of HSPCs in BM. However, all factors that direct mobilization of HSPCs into PB and homing back to the BM or their allocation to damaged organs are not characterized very well. In this chapter we will present mounting evidence that elements of innate immunity such as complement cascade (CC) cleavage fragments (e.g., C3a and C5a), granulocytes, generation of membrane attack complex (MAC) together with sphingosine-1 phosphate (S1P) orchestrate HSPC mobilization. On other hand some other bioactive lipids e.g., ceramide-1-phosphate (C1P) that is released from damaged/"leaky" cells in BM after myeloablative conditioning for transplant may play an opposite important role in homing of HSPCs to BM. Finally, the chemotactic activity of all chemoattractants for HSPCs including SDF-1, S1P and C1P is enhanced in presence of CC cleavage fragments (e.g., C3a) and MAC that is a final product of CC activation.


Assuntos
Quimiotaxia/imunologia , Proteínas do Sistema Complemento/imunologia , Células-Tronco Hematopoéticas/imunologia , Imunidade Inata/imunologia , Esfingolipídeos/imunologia , Animais , Humanos
12.
Differentiation ; 81(3): 153-61, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21339038

RESUMO

From the point of view of regenerative potential, the most important cells are pluripotent stem cells (PSCs). Such cells must fulfill certain in vitro as well as in vivo criteria that have been established by work with PSCs isolated from embryos, which are known as embryonic stem cells (ESCs). According to these criteria, pluripotent stem cells should: (i) give rise to cells from all three germ layers, (ii) complete blastocyst development, and (iii) form teratomas after inoculation into experimental animals. Unfortunately, in contrast to immortalized embryonic ESC lines or induced PSCs (iPSCs), these last two criteria have thus far not been obtained in a reproducible manner for any potential PSC candidates isolated from adult tissues. There are two possible explanations for this failure. The first is that PSCs isolated from adult tissues are not fully pluripotent; the second is that there are some physiological mechanisms involved in keeping these cells quiescent in adult tissues that preclude their "unleashed proliferation", thereby avoiding the risk of teratoma formation. In this review we present an evidence that adult tissues contain remnants from development; a population of PSCs that is deposited in various organs as a backup for primitive stem cells, plays a role in rejuvenation of the pool of more differentiated tissue-committed stem cells (TCSCs), and is involved in organ regeneration. These cells share several markers with epiblast/germ line cells and have been named very small embryonic-like stem cells (VSELs). We suggest that, on one hand, VSELs maintain mammalian life span but, on the other hand, they may give rise to several malignancies if they mutate. We provide an evidence that the quiescent state of these cells in adult tissues, which prevents teratoma formation, is the result of epigenetic changes in some of the imprinted genes.


Assuntos
Células-Tronco Embrionárias/fisiologia , Longevidade , Células-Tronco Pluripotentes/fisiologia , Regeneração , Animais , Biomarcadores/metabolismo , Movimento Celular , Células-Tronco Embrionárias/citologia , Epigênese Genética , Células Germinativas , Humanos , Células-Tronco Pluripotentes/citologia
13.
Leukemia ; 36(1): 248-256, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34285343

RESUMO

Recent evidence indicates that extracellular adenosine triphosphate (eATP), as a major mediator of purinergic signaling, plays an important role in regulating the mobilization and homing of hematopoietic stem progenitor cells (HSPCs). In our previous work we demonstrated that eATP activates the P2X7 ion channel receptor in HSPCs and that its deficiency impairs stem cell trafficking. To learn more about the role of the P2X purinergic receptor family in hematopoiesis, we phenotyped murine and human HSPCs with respect to the seven P2X receptors and observed that, these cells also highly express P2X4 receptors, which shows ~50% sequence similarity to P2X7 subtypes, but that P2X4 cells are more sensitive to eATP and signal much more rapidly. Using the selective P2X4 receptor antagonist PSB12054 as well as P2X4-KO mice, we found that the P2X4 receptor, similar to P2X7 receptor, promotes trafficking of HSPCs in that its deficiency leads to impaired chemotaxis of HSPCs in response to a stromal-derived factor 1 (SDF-1) gradient, less effective pharmacological mobilization, and defective homing and engraftment of HSPCs after transplantation into myeloablated hosts. This correlated with a decrease in SDF-1 expression in the BM microenvironment. Overall, our results confirm the proposed cooperative dependence of both receptors in response to eATP signaling. In G-CSF-induced mobilization, a lack of one receptor is not compensated by the presence of the other one, which supports their mutual dependence in regulating HSPC trafficking.


Assuntos
Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Receptores Purinérgicos P2X4/fisiologia , Receptores Purinérgicos P2X7/metabolismo , Nicho de Células-Tronco , Animais , Quimiotaxia , Feminino , Fator Estimulador de Colônias de Granulócitos/metabolismo , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Purinérgicos P2X7/genética , Transdução de Sinais
14.
J Cell Mol Med ; 15(6): 1319-28, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20629987

RESUMO

Adult bone marrow-derived very small embryonic-like stem cells (VSEL-SCs) exhibit a Sca-1(+)/Lin(-)/CD45(-) phenotype and can differentiate into various cell types, including cardiomyocytes and endothelial cells. We have previously reported that transplantation of a small number (1 × 10(6)) of freshly isolated, non-expanded VSEL-SCs into infarcted mouse hearts resulted in improved left ventricular (LV) function and anatomy. Clinical translation, however, will require large numbers of cells. Because the frequency of VSEL-SCs in the marrow is very low, we examined whether VSEL-SCs can be expanded in culture without loss of therapeutic efficacy. Mice underwent a 30 min. coronary occlusion followed by reperfusion and, 48 hrs later, received an intramyocardial injection of vehicle (group I, n = 11), 1 × 10(5) enhanced green fluorescent protein (EGFP)-labelled expanded untreated VSEL-SCs (group II, n = 7), or 1 × 10(5) EGFP-labelled expanded VSEL-SCs pre-incubated in a cardiogenic medium (group III, n = 8). At 35 days after myocardial infarction (MI), mice treated with pre-incubated VSEL-SCs exhibited better global and regional LV systolic function and less LV hypertrophy compared with vehicle-treated controls. In contrast, transplantation of expanded but untreated VSEL-SCs did not produce appreciable reparative benefits. Scattered EGFP(+) cells expressing α-sarcomeric actin, platelet endothelial cell adhesion molecule (PECAM)-1, or von Willebrand factor were present in VSEL-SC-treated mice, but their numbers were very small. No tumour formation was observed. We conclude that VSEL-SCs expanded in culture retain the ability to alleviate LV dysfunction and remodelling after a reperfused MI provided that they are exposed to a combination of cardiomyogenic growth factors and cytokines prior to transplantation. Counter intuitively, the mechanism whereby such pre-incubation confers therapeutic efficacy does not involve differentiation into new cardiac cells. These results support the potential therapeutic utility of VSEL-SCs for cardiac repair.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células-Tronco Embrionárias , Injeções Intramusculares/métodos , Infarto do Miocárdio/terapia , Miocárdio/patologia , Transplante de Células-Tronco/métodos , Função Ventricular Esquerda , Animais , Medula Óssea/fisiologia , Técnicas de Cultura de Células , Oclusão Coronária/complicações , Meios de Cultura , Citocinas/farmacologia , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Proteínas de Fluorescência Verde/análise , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Remodelação Ventricular
15.
Gerontology ; 57(1): 76-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20134149

RESUMO

Genetic material in the nucleus governs mechanisms related to cell proliferation, differentiation, and function. Thus, senescence and aging are directly tied to the change of nuclear function and structure. The most important mechanisms that affect cell senescence are: (i) telomere shortening; (ii) environmental stress-mediated accumulation of DNA mutations, and (iii) the intrinsically encoded biological clock that dictates lifespan events of any particular cell type. Overall, these changes lead to modification of the expression of genes that are responsible for: (i) organization of the nuclear structure; (ii) integrity of transcriptionally inactive heterochromatin, and (iii) epigenetic modification of chromosomes due to DNA methylation and/or histone modifications. These aging-related nuclear alterations do not only affect somatic cells. More importantly, they affect stem cells, which are responsible for proper tissue rejuvenation. In this review, we focus on epigenetic changes in the chromatin structure and their impact on the biology and function of adult cells as they age. We will also address aging-related changes in a compartment of the most primitive pluripotent stem cells that were recently identified by our team and named 'very small embryonic/epiblast-like stem cells'.


Assuntos
Envelhecimento/genética , Núcleo Celular/genética , Senescência Celular/genética , Montagem e Desmontagem da Cromatina/genética , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Envelhecimento/patologia , Animais , Dano ao DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Impressão Genômica , Humanos , Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Modelos Genéticos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais , Telômero/genética
16.
Stem Cell Rev Rep ; 17(3): 821-828, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33196976

RESUMO

Evidence indicates that bone marrow (BM)-residing hematopoietic stem/progenitor cells (HSPCs) are released into peripheral blood (PB) after administration of pro-mobilizing drugs, which induce a state of sterile inflammation in the BM microenvironment. In the reverse process, as seen after hematopoietic transplantation, intravenously injected HSPCs home and engraft into BM niches. Here again, conditioning for transplantation by myeloablative chemo- or radiotherapy induces a state of sterile inflammation that promotes HSPC seeding to BM stem cell niches. Therefore, the trafficking of HSPCs and their progeny, including granulocytes and monocytes/macrophages, is regulated by a response to pro-inflammatory stimuli. This responsiveness to inflammatory cues is also preserved after malignant transformation of hematopoietic cells. Results from our laboratory indicate that the responsiveness of hematopoietic cells to pro-inflammatory stimuli is orchestrated by Nlrp3 inflammasome. As reported, HO-1 effectively attenuates intracellular activation of Nlrp3 inflammasome as well as the pro-inflammatory effects of several humoral mediators, including complement cascade (ComC) cleavage fragments that promote migration of hematopoietic cells. Based on this finding, inhibition of HO-1 activity may become a practical strategy to enhance the mobilization and homing of normal HSPCs, and, alternatively, its activation may prevent unwanted spread and in vivo expansion of leukemic cells. Graphical Abstract.


Assuntos
Células-Tronco Hematopoéticas , Heme Oxigenase-1 , Medula Óssea/metabolismo , Heme Oxigenase-1/metabolismo , Inflamassomos/metabolismo , Nicho de Células-Tronco
17.
Stem Cell Rev Rep ; 17(1): 266-277, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32691370

RESUMO

Angiotensin-converting enzyme 2 (ACE2) plays an important role as a member of the renin-angiotensin-aldosterone system (RAAS) in regulating the conversion of angiotensin II (Ang II) into angiotensin (1-7) (Ang [1-7]). But at the same time, while expressed on the surface of human cells, ACE2 is the entry receptor for SARS-CoV-2. Expression of this receptor has been described in several types of cells, including hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPCs), which raises a concern that the virus may infect and damage the stem cell compartment. We demonstrate for the first time that ACE2 and the entry-facilitating transmembrane protease TMPRSS2 are expressed on very small CD133+CD34+Lin-CD45- cells in human umbilical cord blood (UCB), which can be specified into functional HSCs and EPCs. The existence of these cells known as very small embryonic-like stem cells (VSELs) has been confirmed by several laboratories, and some of them may correspond to putative postnatal hemangioblasts. Moreover, we demonstrate for the first time that, in human VSELs and HSCs, the interaction of the ACE2 receptor with the SARS-CoV-2 spike protein activates the Nlrp3 inflammasome, which if hyperactivated may lead to cell death by pyroptosis. Based on this finding, there is a possibility that human VSELs residing in adult tissues could be damaged by SARS-CoV-2, with remote effects on tissue/organ regeneration. We also report that ACE2 is expressed on the surface of murine bone marrow-derived VSELs and HSCs, although it is known that murine cells are not infected by SARS-CoV-2. Finally, human and murine VSELs express several RAAS genes, which sheds new light on the role of these genes in the specification of early-development stem cells. Graphical Abstract •Human VSELs and HSCs express ACE2 receptor for SARS-CoV2 entry. •Interaction of viral spike protein with ACE2 receptor may hyperactivate Nlrp3 inflammasome which induces cell death by pyroptosis. •SARS-CoV2 may also enter cells and eliminate them by cell lysis. •What is not shown since these cells express also Ang II receptor they may hyperactivate Nlrp3 inflammasome in response to Ang II which may induce pyroptosis. Our data indicates that Ang 1-7 may have a protective effect.


Assuntos
Enzima de Conversão de Angiotensina 2/genética , COVID-19/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Animais , COVID-19/patologia , COVID-19/virologia , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/virologia , Humanos , Inflamassomos/genética , Antígenos Comuns de Leucócito/genética , SARS-CoV-2/patogenicidade , Serina Endopeptidases/genética , Internalização do Vírus
18.
Leukemia ; 35(9): 2658-2671, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33623143

RESUMO

Like their homing after transplantation to bone marrow (BM), the mobilization of hematopoietic stem/progenitor cells (HSPCs) is still not fully understood, and several overlapping pathways are involved. Several years ago our group proposed that sterile inflammation in the BM microenvironment induced by pro-mobilizing agents is a driving force in this process. In favor of our proposal, both complement cascade (ComC)-deficient and Nlrp3 inflammasome-deficient mice are poor G-CSF and AMD3100 mobilizers. It is also known that the Nlrp3 inflammasome mediates its effects by activating caspase-1, which is responsible for proteolytic activation of interleukin-1ß (IL-1ß) and interleukin-18 (IL-18) and their release from cells along with several danger-associated molecular pattern molecules (DAMPs). We observed in the past that IL-1ß and IL-18 independently promote mobilization of HSPCs. In the current work we demonstrated that caspase-1-KO mice are poor mobilizers, and, to our surprise, administration of IL-1ß or IL-18, as in the case of Nlrp3-KO animals, does not correct this defect. Moreover, neither Caspase-1-KO nor Nlrp3-KO mice properly activated the ComC to execute the mobilization process. Interestingly, mobilization in these animals and activation of the ComC were both restored after injection of the DAMP cocktail eATP+HGMB1+S100A9, the components of which are normally released from cells in an Nlrp3 inflammasome-caspase-1-dependent manner. In addition, we report that caspase-1-deficient HSPCs show a decrease in migration in response to BM homing factors and engraft more poorly after transplantation. These results for the first time identify caspase-1 as an orchestrator of HSPC trafficking.


Assuntos
Alarminas/metabolismo , Caspase 1/fisiologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Inflamação/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Animais , Movimento Celular , Feminino , Fator Estimulador de Colônias de Granulócitos/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/terapia , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
Int J Cancer ; 126(2): 371-81, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19588509

RESUMO

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of adolescence and childhood. Because RMS tumors are highly vascularized, we sought to determine which factors secreted by RMS cells are crucial in stimulating angiogenesis in response to hypoxia. To address this issue, we evaluated expression of several proangiogenic factors [interleukin (IL)-8, vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, stromal-derived factor (SDF)-1, hepatocyte growth factor (HGF) and leukemia inhibitory factor (LIF)] in 8 human RMS cell lines in both normal steady-state and hypoxic conditions. We found by real-time quantitative polymerase chain reaction (RQ-PCR) and confirmed by enzyme-linked immunosorbent assay (ELISA) that from all the factors evaluated, IL-8, whose expression is very low in normoxia, had been very highly expressed and secreted by RMS cells lines during hypoxic conditions ( approximately 40-170 times). Interestingly, this upregulation was not affected by knocking down hypoxia-inducible factor (HIF)-1alpha, but was inhibited by mitogen-activated protein kinase (MAPK)p42/44 and phosphatidylinositaol 3-kinase (PI3K)/AKT pathway inhibitors. This suggests that IL-8 expression is regulated in an activating protein (AP)-1- and nuclear factor (NF)-kappaB-dependent manner. Furthermore, we found that conditioned media (CM) harvested from RMS cells exposed to hypoxia activated and stimulated chemotactic responses in human umbilical vein endothelial cells (HUVECs) and that IL-8 was responsible for hypoxia-related effects. Finally, by employing shRNA, the expression of IL-8 in human RH-30 cells was downregulated. We noticed that such RMS cells, if injected into skeletal muscles of immunodeficient mice, have a reduced ability for tumor formation. We conclude that IL-8 is a pivotal proangiogenic factor released by human RMS cells in hypoxic conditions and that the targeting of IL-8 may prove to be a novel and efficient strategy for inhibiting RMS growth.


Assuntos
Interleucina-8/genética , Rabdomiossarcoma/genética , Regulação para Cima , Animais , Hipóxia Celular , Linhagem Celular , Linhagem Celular Tumoral , Quimiotaxia/efeitos dos fármacos , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Humanos , Hipóxia , Interleucina-8/metabolismo , Camundongos , Camundongos SCID , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Am J Pathol ; 174(6): 1985-92, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19406990

RESUMO

The morphology of several tumors mimics developmentally early tissues, and tumors often express early developmental markers characteristic of the germ line lineage. The presence of these markers in neoplastic cells could reflect the dedifferentiation of somatic cells in which cancer develops or cancer origination in primitive stem cells closely related to the epiblast/germ line. The identification of primitive germ line-derived very small embryonic/epiblast-like stem cells, which are deposited early in embryogenesis in developing organs and persist in several organs into adulthood, raised the possibility that cancer may originate in these cells. In this review, we hypothesize that very small embryonic/epiblast-like stem cells could be a missing link that support the more than 100-year-old concepts of the embryonic rest or germ line origin hypotheses of cancer development; however, further experimental evidence is needed to support this hypothesis.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa