Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
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.
Purinergic Signal ; 16(3): 313-325, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32533388

RESUMO

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.


Assuntos
Trifosfato de Adenosina/metabolismo , Células da Medula Óssea/metabolismo , Conexinas/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Inflamação/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Medula Óssea/metabolismo , Inflamassomos/metabolismo , Camundongos
3.
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
4.
Adv Exp Med Biol ; 1201: 1-22, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31898779

RESUMO

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.


Assuntos
Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Medicina Regenerativa/tendências , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
5.
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
6.
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
8.
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
9.
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
10.
Leukemia ; 38(4): 692-698, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38388648

RESUMO

In single-cell organisms, extracellular microvesicles (ExMVs) were one of the first cell-cell communication platforms that emerged very early during evolution. Multicellular organisms subsequently adapted this mechanism. Evidence indicates that all types of cells secrete these small circular structures surrounded by a lipid membrane that may be encrusted by ligands and receptors interacting with target cells and harboring inside a cargo comprising RNA species, proteins, bioactive lipids, signaling nucleotides, and even entire organelles "hijacked" from the cells of origin. ExMVs are secreted by normal cells and at higher levels by malignant cells, and there are some differences in their cargo. On the one hand, ExMVs secreted from malignant cells interact with cells in the microenvironment, and in return, they are exposed by a "two-way mechanism" to ExMVs secreted by non-leukemic cells. Therefore, leukemogenesis occurs and progresses in ExMVs enriched microenvironments, and this biological fact has pathologic, diagnostic, and therapeutic implications. We are still trying to decipher this intriguing cell-cell communication language better. We will present a current point of view on this topic and review some selected most recent discoveries and papers.


Assuntos
Micropartículas Derivadas de Células , Exossomos , Vesículas Extracelulares , Humanos , Exossomos/metabolismo , Comunicação Celular , Transdução de Sinais , Proteínas/metabolismo , Vesículas Extracelulares/metabolismo
11.
Stem Cell Rev Rep ; 20(3): 827-838, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38363476

RESUMO

A cell's most significant existential task is to survive by ensuring proper metabolism, avoiding harmful stimuli, and adapting to changing environments. It explains why early evolutionary primordial signals and pathways remained active and regulate cell and tissue integrity. This requires energy supply and a balanced redox state. To meet these requirements, the universal intracellular energy transporter purine nucleotide-adenosine triphosphate (ATP) became an important signaling molecule and precursor of purinergic signaling after being released into extracellular space. Similarly, ancient proteins involved in intracellular metabolism gave rise to the third protein component (C3) of the complement cascade (ComC), a soluble arm of innate immunity. These pathways induce cytosol reactive oxygen (ROS) and reactive nitrogen species (RNS) that regulate the redox state of the cells. While low levels of ROS and RNS promote cell growth and differentiation, supra-physiological concentrations can lead to cell damage by pyroptosis. This balance explains the impact of purinergic signaling and innate immunity on cell metabolism, organogenesis, and tissue development. Subsequently, along with evolution, new regulatory cues emerge in the form of growth factors, cytokines, chemokines, and bioactive lipids. However, their expression is still modulated by both primordial signaling pathways. This review will focus on the data that purinergic signaling and innate immunity carry on their ancient developmental task in hematopoiesis and specification of hematopoietic stem/progenitor cells (HSPCs). Moreover, recent evidence shows both these regulatory pathways operate in a paracrine manner and inside HSPCs at the autocrine level.


Assuntos
Células-Tronco Hematopoéticas , Imunidade Inata , Espécies Reativas de Oxigênio/metabolismo , Ativação do Complemento , Hematopoese
12.
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
13.
Prostaglandins Other Lipid Mediat ; 104-105: 122-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22981511

RESUMO

Under steady-state conditions, hematopoietic stem/progenitor cells (HSPCs) egress from bone marrow (BM) and enter peripheral blood (PB) where they circulate at low levels. Their number in PB, however, increases significantly in several stress situations related to infection, organ/tissue damage, or strenuous exercise. Pharmacologically mediated enforced egress of HSPCs from the BM microenvironment into PB is called "mobilization", and this phenomenon has been exploited in hematological transplantology as a means to obtain HSPCs for hematopoietic reconstitution. In this review we will present the accumulated evidence that innate immunity, including the complement cascade and the granulocyte/monocyte lineage, and the PB plasma level of the bioactive lipid sphingosine-1-phosphate (S1P) together orchestrate this evolutionarily conserved mechanism that directs trafficking of HSPCs.


Assuntos
Anemia Falciforme/metabolismo , Proteínas do Sistema Complemento/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Anemia Falciforme/imunologia , Anemia Falciforme/patologia , Quimiotaxia/imunologia , Ativação do Complemento , Proteínas do Sistema Complemento/imunologia , Granulócitos/imunologia , Granulócitos/metabolismo , Granulócitos/patologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Hemoglobinúria Paroxística/imunologia , Hemoglobinúria Paroxística/patologia , Humanos , Imunidade Inata , Lisofosfolipídeos/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Esfingosina/imunologia , Esfingosina/metabolismo
14.
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
15.
Adv Exp Med Biol ; 735: 219-32, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23402030

RESUMO

The first step that precedes hematopoietic transplantation is elimination of pathological hematopoiesis by administration of myeloablative doses of radiochemotherapy. This eliminates hematolymphopoietic cells and at the same time damages hematopoietic microenvironment in bone marrow (BM). The damage of BM tissue leads to activation of complement cascade (CC), and bioactive CC cleavage fragments modulate several steps of BM recovery after transplantation of hematopoietic stem progenitor cells (HSPCs). Accordingly, C3 cleavage fragments (soluble C3a/desArgC3a and solid phase iC3b) and generation of soluble form of C5b-C9 also known as membrane attack complex (MAC) as well as release of antimicrobial cationic peptides from stromal cells (cathelicidin or LL-37 and beta-2 defensin) promote homing of HSPCs. To support this, C3 cleavage fragments and antimicrobial cationic peptides increase homing responsiveness of transplanted HSPCs to stroma-derived factor-1 (SDF-1) gradient. Furthermore, damaged BM cells release several other chemoattractants for HSPCs such as bioactive lipids sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P) and chemotactic purines (ATP and UTP). In this chapter, we will discuss the current view on homing of transplanted HSPCs into BM that in addition to SDF-1 is orchestrated by CC, antimicrobial cationic peptides, and several other prohoming factors. We also propose modulation of CC as a novel strategy to optimize/accelerate homing of HSPCs.


Assuntos
Medula Óssea/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Imunidade Inata/fisiologia , Animais , Peptídeos Catiônicos Antimicrobianos/fisiologia , Quimiocina CXCL12/fisiologia , Proteínas do Sistema Complemento/fisiologia , Humanos , Lipídeos/fisiologia , beta-Defensinas/fisiologia , Catelicidinas
16.
Stem Cell Rev Rep ; 19(5): 1177-1184, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36976465

RESUMO

Hematopoietic stem/progenitor cells (HSPCs) express receptors for complement cascade (ComC) cleavage fragments C3a and C5a and may respond to inflammation-related cues by sensing pathogen-associated molecular pattern molecules (PAMPs) released by pathogens as well as non-infectious danger associated molecular pattern molecules (DAMPs) or alarmin generated during stress/tissue damage sterile inflammation. To facilitate this HSPCs are equipped with C3a and C5a receptors, C3aR and C5aR, respectively, and express on the outer cell membrane and in cytosol pattern recognition receptors (PPRs) that sense PAMPs and DAMPs. Overall, danger-sensing mechanisms in HSPCs mimic those seen in immune cells, which should not surprise as hematopoiesis and the immune system develop from the same common stem cell precursor. This review will focus on the role of ComC-derived C3a and C5a that trigger nitric oxide synthetase-2 (Nox2) complex to release reactive oxygen species (ROS) that activate important cytosolic PRRs-Nlrp3 inflammasome, which orchestrates responsiveness of HSPCs to stress. Moreover, recent data indicate that in addition to circulating in peripheral blood (PB) activated liver-derived ComC proteins, a similar role plays ComC expressed and intrinsically activated in HSPCs known as "complosome". We postulate that ComC triggered Nox2-ROS-Nlrp3 inflammasome responses, if they occur within non-toxic to cells' "hormetic range of activation", positively regulate HSCs migration, metabolism, and proliferation. This sheds a new light on the immune-metabolic regulation of hematopoiesis.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Humanos , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Proteínas do Sistema Complemento/metabolismo , Inflamação/metabolismo , Fígado/metabolismo
17.
Stem Cell Rev Rep ; 19(7): 2292-2298, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37386334

RESUMO

Bone marrow (BM) contains not only hematopoietic stem cells (HSCs) but also some very rare, early development, small quiescent stem cells that, upon activation, may differentiate across germlines. These small cells, named very small embryonic like stem cells (VSELs), can undergo specification into several types of cells including HSCs. Interestingly, murine BM is also home to a "mystery" population of small CD45+ stem cells with many of the phenotypic characteristics attributed to resting HSCs. Since the size of the "mystery" population cells are between that of VSELs and HSCs, and because CD45- VSELs can be specified into CD45+ HSCs, we hypothesized that the quiescent CD45+ "mystery" population could be a missing developmental link between VSELs and HSCs. To support this hypothesis, we showed that VSELs first became enriched for HSCs after acquiring expression of the CD45 antigen already expressed on "mystery" stem cells. Moreover, VSELs freshly isolated from BM similar to the "mystery" population cells, are quiescent and do not reveal hematopoietic potential in in vitro and in vivo assays. However, we noticed that CD45+ "mystery" population cells, similar to CD45- VSELs, became specified into HSCs after co-culture over OP9 stroma. We also found that mRNA for Oct-4, a pluripotency marker that is highly expressed in VSELs, is also detectable in the "mystery" population cells, albeit at a much lower level. Finally, we determined that the "mystery" population cells specified over OP9 stroma support were able to engraft and establish hematopoietic chimerism in lethally irradiated recipients. Based on these results, we propose that the murine BM "mystery" population could be an intermediate population between BM-residing VSELs and HSCs already specified for lympho-hematopoietic lineages.


Assuntos
Medula Óssea , Células-Tronco Hematopoéticas , Camundongos , Animais , Diferenciação Celular , Células-Tronco Embrionárias , Células da Medula Óssea
18.
Stem Cell Rev Rep ; 19(1): 92-103, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36441489

RESUMO

Proliferation, metabolism, and migration of hematopoietic stem/progenitor cells (HSPCs) are coordinated by receptors expressed on outer cell membranes that are integrated into microdomains, known as membrane lipid rafts (MLRs). These structures float freely in the cell membrane bilayer and are enriched in cholesterol and sphingolipids for their functional integrity. Receptors, if expressed in MLRs, have prolonged occupancy on the cell surface and enhanced signaling power. Based on this, we have become interested in the regulation of synthesis of MLRs components in HSPCs. To address this, we tested the effect of selected factors that promote proliferation or migration and their potential involvement in the synthesis of MLRs components in HSPCs. Based on our previous research showing that HSPCs from Nox2-KO and Nlrp3-KO mice display a profound defect in MLRs formation, we focused on the role of Nox2-ROS-Nlrp3 inflammasome in regulating lipogenesis in HSPCs. We found that while at steady state conditions, Nox2-derived ROS is required for a proper expression of enzymes regulating lipogenesis, during inflammation, this effect is augmented by Nlrp3 inflammasome. Thus, our data sheds new light on the regulation of lipogenesis in HSPCs and the involvement of the Nox2-ROS-Nlrp3 inflammasome axis that differently regulates lipogenesis at steady state conditions and in response to inflammation, modulating MLRs-mediated responsiveness of these cells to external stimuli.


Assuntos
Inflamassomos , Lipogênese , Animais , Camundongos , Inflamassomos/metabolismo , Lipogênese/genética , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco Hematopoéticas , Inflamação/metabolismo , Lipídeos de Membrana/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
19.
Methods Mol Biol ; 2567: 263-280, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36255707

RESUMO

Mobilization or egress of stem cells from bone marrow (BM) into peripheral blood (PB) is an evolutionary preserved and important mechanism in an organism for self-defense and regeneration. BM-derived stem cells circulate always at steady-state conditions in PB, and their number increases during stress situations related to (a) infections, (b) tissue organ injury, (c) stress, and (d) strenuous exercise. Stem cells also show a circadian pattern of their PB circulating level with peak in early morning hours and nadir late at night. The number of circulating in PB stem cells could be pharmacologically increased after administration of some drugs such as cytokine granulocyte colony-stimulating factor (G-CSF) or small molecular antagonist of CXCR4 receptor AMD3100 (Plerixafor) that promote their egress from BM into PB and lymphatic vessels. Circulating can be isolated from PB for transplantation purposes by leukapheresis. This important homeostatic mechanism is governed by several intrinsic complementary pathways. In this chapter, we will discuss the role of purinergic signaling and extracellular nucleotides in regulating this process and review experimental strategies to study their involvement in mobilization of various types of stem cells that reside in murine BM.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Compostos Heterocíclicos , Camundongos , Animais , Receptores CXCR4/metabolismo , Compostos Heterocíclicos/farmacologia , Fator Estimulador de Colônias de Granulócitos , Células da Medula Óssea/metabolismo , Nucleotídeos
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa