Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Opt Express ; 24(25): 28877-28888, 2016 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-27958553

RESUMO

An experimental setup capable of measuring simultaneous 2D scattered light angular distribution from two directions to study cell morphology without the use of bio-labels was developed. Experiments with hematopoietic stem cells (CD34+ cells) show good agreement with detailed numerical simulations of light scattering. Numerical simulations and computer models of cells are used to identify physical features of cells with the largest scattering cross sections. This allows for determination of size, geometry of the nucleus and distribution of mitochondria in hematopoietic stem cells by means of our label-free method.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Óptica e Fotônica , Contagem de Células , Núcleo Celular , Células-Tronco Hematopoéticas/classificação , Luz
2.
Oncotarget ; 7(3): 3033-46, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26701888

RESUMO

We recently reported that normal hematopoietic stem cells express functional pituitary sex hormone (SexH) receptors. Here we report for the first time that pituitary-secreted gonadotrophins stimulate migration, adhesion, and proliferation of several human myeloid and lymphoid leukemia cell lines. Similar effects were observed after stimulation of human leukemic cell lines by gonadal SexHs. This effect seems to be direct, as the SexH receptors expressed by leukemic cells responded to stimulation by phosphorylation of MAPKp42/44 and AKTser473. Furthermore, in parallel studies we confirmed that human primary patient-derived AML and CML blasts also express several functional SexH receptors. These results shed more light on the potential role of SexHs in leukemogenesis and, in addition, provide further evidence suggesting a developmental link between hematopoiesis and the germline.


Assuntos
Hormônio Foliculoestimulante/farmacologia , Gonadotropinas Hipofisárias/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mieloide Aguda/metabolismo , Hormônio Luteinizante/farmacologia , Receptores do Hormônio Hipofisário/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Células Jurkat , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Mieloides/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
3.
Cryobiology ; 71(2): 181-97, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26186998

RESUMO

Originally isolated from bone marrow, mesenchymal stromal cells (MSCs) have since been obtained from various fetal and post-natal tissues and are the focus of an increasing number of clinical trials. Because of their tremendous potential for cellular therapy, regenerative medicine and tissue engineering, it is desirable to cryopreserve and bank MSCs to increase their access and availability. A remarkable amount of research and resources have been expended towards optimizing the protocols, freezing media composition, cooling devices and storage containers, as well as developing good manufacturing practices in order to ensure that MSCs retain their therapeutic characteristics following cryopreservation and that they are safe for clinical use. Here, we first present an overview of the identification of MSCs, their tissue sources and the properties that render them suitable as a cellular therapeutic. Next, we discuss the responses of cells during freezing and focus on the traditional and novel approaches used to cryopreserve MSCs. We conclude that viable MSCs from diverse tissues can be recovered after cryopreservation using a variety of freezing protocols, cryoprotectants, storage periods and temperatures. However, alterations in certain functions of MSCs following cryopreservation warrant future investigations on the recovery of cells post-thaw followed by expansion of functional cells in order to achieve their full therapeutic potential.


Assuntos
Criopreservação/métodos , Crioprotetores/farmacologia , Células-Tronco Mesenquimais/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Congelamento , Humanos , Engenharia Tecidual/métodos , Vitrificação
4.
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
5.
Mol Ther Nucleic Acids ; 4: e240, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25965550

RESUMO

Leukemias arise from genetic alterations in normal hematopoietic stem or progenitor cells, leading to impaired regulation of proliferation, differentiation, apoptosis, and survival of the transformed cells. With the advent of RNA interference (RNAi) and the short interfering RNA (siRNA) as its pharmacological mediator, it is becoming possible to modulate specific targets at will. This article summarizes current attempts to utilize RNAi reagents for therapy of leukemias, focusing on acute and chronic myeloid leukemia. We first present unique aspects of RNAi-mediated therapy, followed by a brief background on the delivery technology of RNAi reagents. The need for leukemia-specific delivery of siRNA is discussed by describing approaches that targeted agents to leukemic cells. Pharmacokinetics and biodistribution of RNAi agents are then presented, highlighting the critical issues pertinent to emerging siRNA therapy. Efforts to deliver specific RNAi therapies are then summarized in the context of expected clinical outcomes, focusing on limiting leukemic cell survival, sensitizing malignant cells to chemotherapy, mobilization of leukemic cells, and eradication of leukemic stem cells. We conclude with a perspective on the future of RNAi therapy, emphasizing the technological requirements and mechanistic challenges for clinical entry.

6.
Artif Cells Nanomed Biotechnol ; 43(5): 318-27, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24564349

RESUMO

Continuous delivery of proteins by engineered cells encapsu-lated in biocompatible polymeric microcapsules is of considerable therapeutic potential. However, this technology has not lived up to expectations due to inadequate cell--matrix interactions and subsequent cell death. In this study we hypoth-esize that the presence of fibronectin in an alginate matrix may enhance the viability and functionality of encapsulated human cord blood-derived mesenchymal stromal cells (MSCs) expressing the human Factor IX (FIX) gene. MSCs were encapsulated in alginate-PLL microcapsules containing 10, 100, or 500 µg/ml fibronectin to ameliorate cell survival. MSCs in microcapsules with 100 and 500 µg/ml fibronectin demonstrated improved cell viability and proliferation and higher FIX secretion compared to MSCs in non-supplemented microcapsules. In contrast, 10 µg/ml fibronectin did not significantly affect the viability and protein secretion from the encapsulated cells. Differentiation studies demonstrated osteogenic (but not chondrogenic or adipogenic) differentiation capability and efficient FIX secretion of the enclosed MSCs in the fibronectin-alginate suspension culture. Thus, the use of recombinant MSCs encapsulated in fibronectin-alginate microcapsules in basal or osteogenic cultures may be of practical use in the treatment of hemophilia B.


Assuntos
Alginatos/química , Engenharia Celular , Fator IX/metabolismo , Fibronectinas/química , Células-Tronco Mesenquimais/metabolismo , Cápsulas , Sobrevivência Celular , Células Cultivadas , Células Imobilizadas/citologia , Células Imobilizadas/metabolismo , Fator IX/genética , Ácido Glucurônico/química , Ácidos Hexurônicos/química , Humanos , Células-Tronco Mesenquimais/citologia
7.
Leuk Res ; 38(11): 1299-308, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25262448

RESUMO

The adhesion receptor CD44 plays an important role in the survival and retention of leukemic stem/progenitor cells (LSPC) within the bone marrow (BM) niche, as well as in the high relapse rates of acute myeloid leukemia (AML). Down-regulating CD44 could be clinically relevant not only for suppression of the deregulated function of LSPC but also in LSPC response to chemotherapeutic agents. Small interfering RNA (siRNA) delivery is a promising approach for AML treatment, and we recently reported effective siRNA delivery into difficult-to-transfect AML cell lines using lipid-substituted polyethylenimine/siRNA complexes (polymeric nanoparticles). In this study, we investigated polymeric nanoparticle-mediated silencing of CD44 in CD34+ LSPC cell models (leukemic KG-1 and KG-1a cell lines) as well as primary AML cells. Polymeric nanoparticle-mediated silencing decreased surface CD44 levels in KG-1, KG-1a and primary AML cells by up to 27%, 30% and 20% at day 3, respectively. Moreover, CD44 silencing resulted in induction of apoptosis in KG-1 cells, reduced adhesion of KG-1 and KG-1a cells to hyaluronic acid-coated cell culture plates and BM-MSC, and decreased adhesion of primary AML cells to BM-MSC. Our results suggest that polymeric nanoparticle-mediated silencing of CD44 might be a useful technique for inhibiting LSPC interactions with their microenvironment, thereby prohibiting leukemia progression or sensitizing LSPC to chemotherapy.


Assuntos
Antígenos CD34/imunologia , Inativação Gênica , Receptores de Hialuronatos/metabolismo , Leucemia Mieloide Aguda/imunologia , Nanopartículas , Polímeros/química , Receptores de Antígenos/genética , Diferenciação Celular , Linhagem Celular Tumoral , Humanos , Leucemia Mieloide Aguda/patologia , Reação em Cadeia da Polimerase , RNA Interferente Pequeno
8.
J Gene Med ; 16(5-6): 131-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24947827

RESUMO

BACKGROUND: Hemophilia B patients are subject to frequent and spontaneous bleeding caused by a deficiency of clotting factor IX (FIX). Mesenchymal stromal cells (MSCs) have been used in cellular therapies as a result of their immunomodulatory properties, the ability to home to sites of injury and their amenability to various ex vivo modifications, including lentiviral-mediated gene transfer. METHODS: MSCs were isolated from human umbilical cord blood and differentiated into adipogenic, chondrogenic and osteogenic lineages. A lentiviral DNA vector containing the human FIX gene was generated using traditional restriction enzyme digest and ligation techniques to generate viable replication-incompetent lentiviral particles that were used to transduce MSCs. Quantitative measurement of FIX expression was conducted using an enzyme-linked immunosorbent assay. RESULTS: The over-expression of FIX was sustained in vitro at levels > 4 µg/10(6) cells/24 h and FIX coagulant activity was > 2.5 mIU/10(6) cells/24 h for the 6-week duration of study. Lentiviral modification of cells with a multiplicity of infection of 10 did not adversely affect the potential of cord blood (CB) MSCs to differentiate to adipocytes, chondrocytes and osteoblastic cells, and the expression of functional FIX was sustained after differentiation and was similar to that in nondifferentiated cells. CONCLUSIONS: Modification of human CB MSCs with a lentiviral vector resulted in sustained high FIX expression in vitro after differentiation to adipogenic, chondrogenic and osteoblastic cells. These modified MSCs could have applications in cellular therapies for hemophilia B.


Assuntos
Fator IX/genética , Sangue Fetal/citologia , Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Adipogenia , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Condrogênese , Fator IX/metabolismo , Ordem dos Genes , Vetores Genéticos/genética , Hemofilia B/genética , Hemofilia B/terapia , Humanos , Lentivirus/genética , Células-Tronco Mesenquimais/citologia , Camundongos , Tempo de Tromboplastina Parcial , Transdução Genética
9.
Stem Cells Int ; 2014: 610495, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24757448

RESUMO

Mesenchymal stromal cells (MSC) have great potential for cellular therapies as they can be directed to differentiate into certain lineages or to exert paracrine effects at sites of injury. The interactions between stromal cell-derived factor (SDF)-1 and its receptors CXCR4 and CXCR7 play pivotal roles in the migration of MSC to injured tissues. We evaluated whether a histone deacetylase inhibitor valproic acid (VPA) modulates the migration of cord blood (CB-) derived MSC towards SDF-1 and their proliferation and differentiation. We found that in MSC, VPA increased (i) the gene and total protein expression of CXCR4 and CXCR7 and primed migration towards a low gradient of SDF-1, (ii) the gene expression of MMP-2 and secretion and activation of proMMP-2, (iii) the proliferation and gene expression of pluripotency markers SOX2 and Oct-4, and exposure to lower concentrations of VPA (≤5 mM) had no effect on their differentiation to osteocytes and chondrocytes. Thus, our study indicates that VPA enhances the migration of CB MSC towards SDF-1 by increasing the expression of CXCR4, CXCR7, and MMP-2. VPA at low concentrations may be used for ex vivo treatment of MSC to increase their recruitment to sites of injury without compromising their ability to proliferate or differentiate.

10.
Artif Cells Nanomed Biotechnol ; 42(2): 102-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23802745

RESUMO

The success of cell microencapsulation technology in tissue engineering and protein delivery applications depends on the viability and functionality of the encapsulated cells, which in turn are dependent upon cell/matrix interactions. In this work, we compared the viability of cord blood-derived mesenchymal stromal cells (CB MSCs), engineered to secrete factor IX (FIX) for hemophilia treatment, and encapsulated in arginine-glycine-aspartate (RGD)-alginate versus fibrinogen-alginate microcapsules. We evaluated the effect of the biomimetic matrix on cell attachment, proliferation, and secretion of FIX. Compared with nonsupplemented alginate matrix, RGD-alginate significantly enhanced the viability of the encapsulated MSCs. Further, cells in RGD-alginate displayed distinct attachment morphology, thus suggesting that RGD-alginate can potentially be used for the encapsulation of MSCs in tissue engineering applications that require enhanced cell attachment and viability. However, our data also showed that RGD-alginate microcapsules, in contrast to fibrinogen-alginate microcapsules, did not significantly improve cell proliferation of or FIX secretion by encapsulated MSCs. Our findings suggest that evidence of cell attachment alone may not accurately predict the functionality of cells in biomimetic microcapsules.


Assuntos
Cápsulas/metabolismo , Fator IX/metabolismo , Sangue Fetal/fisiologia , Hemofilia A/terapia , Células-Tronco Mesenquimais/fisiologia , Alginatos/química , Cápsulas/química , Adesão Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Composição de Medicamentos/métodos , Matriz Extracelular/metabolismo , Fator IX/genética , Fibrinogênio/química , Terapia Genética , Ácido Glucurônico/química , Hemofilia A/genética , Ácidos Hexurônicos/química , Humanos , Oligopeptídeos/química , Engenharia Tecidual
11.
Cytotherapy ; 15(7): 840-9, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23623275

RESUMO

BACKGROUND AIMS: The interaction between stromal cell-derived factor (SDF)-1 and its receptor CXCR4 is one of the mechanisms by which mesenchymal stromal cells (MSCs) are recruited to sites of injury. SDF-1 is upregulated in damaged tissues, but because the surface expression of CXCR4 on cultured MSCs is low, we investigated whether the delivery of CXCR4 into MSCs with the use of the cationic liposomal reagent IBAfect would increase their migration toward SDF-1. METHODS: We examined (i) the effect of MSC confluency, passage number, duration of transfection and amount of IBAfect and plasmid on transfection efficiency as determined by flow cytometric analysis of CXCR4 and (ii) whether IBAfect-mediated CXCR4 transfection affected the viability, proliferation and differentiation of MSCs as well as their response toward an SDF-1 gradient in a trans-Matrigel migration assay. RESULTS: We found that transfection efficiency of up to 40% was achieved after 24-h transfection of 50% confluent MSCs (at passage 4) with an IBAfect:plasmid ratio of 3.6 µL:0.6 µg, and CXCR4 transcript expression in transfected MSCs was 10(5)-fold higher than in non-transfected cells. Transfected MSCs retained their ability to differentiate to osteocytes and chondrocytes but had lower proliferation. Importantly, overexpression of surface CXCR4 with the use of IBAfect significantly increased (>3-fold) the number of cells migrating toward an SDF-1 gradient relative to cells migrating to media alone, compared with non-transfected cells (1.3-fold). CONCLUSIONS: Our results suggest that IBAfect-mediated delivery of CXCR4 into MSCs is a highly efficient technique that may be useful for enhancing the recruitment of systemically infused MSCs for tissue repair.


Assuntos
Quimiocina CXCL12/metabolismo , Sangue Fetal/citologia , Células-Tronco Mesenquimais/citologia , Receptores CXCR4/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Condrócitos/citologia , Sangue Fetal/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Lipossomos/administração & dosagem , Lipossomos/química , Células-Tronco Mesenquimais/metabolismo , Osteócitos/citologia , Receptores CXCR4/administração & dosagem , Receptores CXCR4/genética , Transfecção , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
12.
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
13.
Biomed Res Int ; 2013: 561098, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24381939

RESUMO

Mesenchymal stromal cells (MSCs) are currently being investigated in numerous clinical trials of tissue repair and various immunological disorders based on their ability to secrete trophic factors and to modulate inflammatory responses. MSCs have been shown to migrate to sites of injury and inflammation in response to soluble mediators including the chemokine stromal cell-derived factor-(SDF-)1, but during in vitro culture expansion MSCs lose surface expression of key homing receptors particularly of the SDF-1 receptor, CXCR4. Here we review studies on enhancement of SDF-1-directed migration of MSCs with the premise that their improved recruitment could translate to therapeutic benefits. We describe our studies on approaches to increase the CXCR4 expression in in vitro-expanded cord blood-derived MSCs, namely, transfection, using the commercial liposomal reagent IBAfect, chemical treatment with the histone deacetylase inhibitor valproic acid, and exposure to recombinant complement component C1q. These methodologies will be presented in the context of other cell targeting and delivery strategies that exploit pathways involved in MSC migration. Taken together, these findings indicate that MSCs can be manipulated in vitro to enhance their in vivo recruitment and efficacy for tissue repair.


Assuntos
Movimento Celular/genética , Quimiocina CXCL12/biossíntese , Células-Tronco Mesenquimais/citologia , Receptores CXCR4/biossíntese , Diferenciação Celular , Quimiocina CXCL12/genética , Sangue Fetal/citologia , Humanos , Receptores CXCR4/genética , Transdução de Sinais/genética
14.
Biotechnol Lett ; 34(10): 1965-73, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22763849

RESUMO

Low-intensity pulsed ultrasound (LIPUS) stimulated the viability, proliferation and differentiation of hematopoietic stem/progenitor cells (HSPC) from fresh and cryopreserved peripheral blood leukapheresis product, as well as cord blood when applied for 10 min each day for 4 days. Cell viability, proliferation and differentiation were assessed on day 5 by viable cell counting, MTS proliferation assay, flow cytometry, and colony-forming unit assay. LIPUS stimulation: (i) enhanced the proliferation of fresh HSPC and maintained the viability of cryopreserved HSPC in vitro; (ii) did not affect the percentage of CD34(+) and CD14(+) cells; and (iii) enhanced burst-forming unit-erythroid colony formation. Hence, we suggest that this novel LIPUS stimulation approach might enhance the efficacy of clinical transplantation and cellular therapies using HSPC.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos da radiação , Som , Antígenos CD34/metabolismo , Diferenciação Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Sangue Fetal/citologia , Humanos , Leucaférese , Receptores de Lipopolissacarídeos/metabolismo , Projetos de Pesquisa
15.
ScientificWorldJournal ; 2012: 758512, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701372

RESUMO

The α-chemokine stromal derived factor 1 (SDF-1), which binds to the CXCR4 and CXCR7 receptors, directs migration and homing of CXCR4+ hematopoietic stem/progenitor cells (HSPCs) to bone marrow (BM) and plays a crucial role in retention of these cells in stem cell niches. However, this unique role of SDF-1 has been recently challenged by several observations supporting SDF-1-CXCR4-independent BM homing. Specifically, it has been demonstrated that HSPCs respond robustly to some bioactive lipids, such as sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), and migrate in response to gradients of certain extracellular nucleotides, including uridine triphosphate (UTP) and adenosine triphosphate (ATP). Moreover, the responsiveness of HSPCs to an SDF-1 gradient is enhanced by some elements of innate immunity (e.g., C3 complement cascade cleavage fragments and antimicrobial cationic peptides, such as cathelicidin/LL-37 or ß2-defensin) as well as prostaglandin E2 (PGE2). Since all these factors are upregulated in BM after myeloblative conditioning for transplantation, a more complex picture of homing emerges that involves several factors supporting, and in some situations even replacing, the SDF-1-CXCR4 axis.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Humanos
16.
Biochem Res Int ; 2012: 685267, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496978

RESUMO

Hematopoietic stem/progenitor cells (HSPCs) are used in clinical transplantation to restore hematopoietic function. Here we review the role of the soluble matrix metalloproteinases MMP-2 and MMP-9, and membrane type (MT)1-MMP in modulating processes critical to successful transplantation of HSPC, such as mobilization and homing. Growth factors and cytokines which are employed as mobilizing agents upregulate MMP-2 and MMP-9. Recently we demonstrated that MT1-MMP enhances HSPC migration across reconstituted basement membrane, activates proMMP-2, and contributes to a highly proteolytic bone marrow microenvironment that facilitates egress of HSPC. On the other hand, we reported that molecules secreted during HSPC mobilization and collection, such as hyaluronic acid and thrombin, increase MT1-MMP expression in cord blood HSPC and enhance (prime) their homing-related responses. We suggest that modulation of MMP-2, MMP-9, and MT1-MMP expression has potential for development of new therapies for more efficient mobilization, homing, and engraftment of HSPC, which could lead to improved transplantation outcomes.

18.
Cytotherapy ; 14(3): 285-95, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22264191

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSC) have great potential for tissue regeneration and cellular therapy. They migrate preferentially to sites of inflammation and tissue injury, but the molecular signals that guide them to their target tissue remain to be elucidated. We have shown that complement component 1 subcomponent q (C1q) enhances the homing-related response of hematopoietic stem/progenitor cells. METHODS: In this study, we investigated whether C1q elicits directional signals that could influence the migration of MSC to injured tissues/organs. RESULTS: We found that C1q chemoattracted human umbilical cord blood-derived MSC in a dose-dependent manner and that the receptor for the global domains of Clq (gC1qR) is present on the surface of MSC. Specific gC1qR antibody blocked the chemotactic response of MSC to C1q, indicating that the C1q/gC1qR interaction may be responsible for the C1q-mediated migration of MSC. Further, we found that C1q enhanced/primed the migration of MSC across reconstituted basement membrane Matrigel towards a low gradient of the chemokine stromal cell-derived factor-1 (SDF-1), which is also present at sites of injury, partly as a result of an increase in surface expression of the SDF-1 receptor CXCR4. Moreover, C1q increased the secretion by MSC of matrix metalloproteinase (MMP)-2 and induced the phosphorylation of ERK1/2. CONCLUSIONS: These results indicate that C1q mediates the migration of MSC in two ways: first, by acting as a chemoattractant, and second, by priming chemotactic responses to SDF-1. Our findings suggest new molecular mechanisms of MSC migration that may contribute to their clinical application in tissue repair.


Assuntos
Movimento Celular , Complemento C1q/farmacologia , Sangue Fetal/citologia , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Fatores Quimiotáticos/metabolismo , Fatores Quimiotáticos/farmacologia , Quimiotaxia , Colágeno/metabolismo , Complemento C1q/metabolismo , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Laminina/metabolismo , Sistema de Sinalização das MAP Quinases , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fosforilação , Proteoglicanas/metabolismo , Receptores de Superfície Celular/metabolismo
19.
Stem Cells Dev ; 21(10): 1587-96, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22047530

RESUMO

The chemokine stromal cell-derived factor (SDF)-1α/CXCL12 and its receptor CXC chemokine receptor 4 (CXCR4) play a crucial role in the homing/engraftment and retention of hematopoietic stem/progenitor cells (HSPCs) in the bone marrow. It has been shown using the viral gene transfer technique that CXCR4 overexpression on human CD34(+) HSPC significantly improves their engraftment in murine models. However, clinical trials with gene therapy have revealed safety concerns related to the immunogenicity of the viral carriers, due to the random integration of viral genes into the host genome. Therefore, a method for CXCR4 gene delivery into HSPC that is safe, nonviral, and highly efficient is needed to improve clinical transplantation and gene therapies. In this work, we investigated the nonviral CXCR4 gene delivery into HSPC using the cationic liposome agent IBAfect. We used CD34(+) cells from cord blood and the models of immature hematopoietic cells expressing CD34 antigen, namely, leukemic cell lines KG-1a and KG-1. Transfection efficiency was determined by flow cytometric analysis 12, 24, 48, and 72 h after transfection, and the viability of cells analyzed by trypan blue exclusion and MTS assays. The functional response of CXCR4-transfected HSPC toward an SDF-1α gradient was determined by chemotaxis assay. We found that ~25% transfection is achieved for KG-1a and KG-1 cells and 20% for HSPC, and that the viability of CXCR4-transfected HSPC is not significantly altered. More importantly, overexpression of CXCR4 using IBAfect significantly increased the chemotaxis of KG-1 cells and HSPC toward SDF-1α. However, we tested 2 other commercially available cationic liposomes (Lipofectamine 2000 and 1,2-dioleoyl-3-trimethylammonium-propane [DOTAP]) in parallel, and we found that they failed to deliver the CXCR4 gene into cells under the same conditions. These results suggest that IBAfect-mediated in vitro gene delivery to overexpress CXCR4 on HSPC is a safe and efficient technique with great potential for improving the efficacy of HSPC transplantation and gene therapy protocols.


Assuntos
Terapia Genética , Células-Tronco Hematopoéticas/fisiologia , Receptores CXCR4/genética , Transfecção , Antígenos CD34/metabolismo , Sobrevivência Celular , Células Cultivadas , Quimiocina CXCL12/química , Quimiotaxia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Lipossomos , Receptores CXCR4/biossíntese , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Estatísticas não Paramétricas
20.
Cancers (Basel) ; 4(3): 743-62, 2012 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-24213464

RESUMO

Membrane type-1 matrix metalloproteinase (MT1-MMP) has been implicated in tumor invasion, as well as trafficking of normal hematopoietic cells, and acts as a physiologic activator of proMMP-2. In this study we examined MT1-MMP expression in primary acute myeloid leukemia (AML) cells. Because tumor necrosis factor (TNF)-α is known to be elevated in AML, we also investigated the effect of TNF-α on MT1-MMP expression. We found (i) MT1-MMP mRNA expression in 41 out of 43 primary AML samples tested; (ii) activation of proMMP-2 in co-cultures of AML cells with normal bone marrow stromal cells; and (iii) inhibition of proMMP-2 activation and trans-Matrigel migration of AML cells by gene silencing using MT1-MMP siRNA. Moreover, recombinant human TNF-α upregulated MT1-MMP expression in AML cells resulting in enhanced proMMP-2 activation and trans-Matrigel migration. Thus, AML cells express MT1-MMP and TNF-α enhances it leading to increased MMP-2 activation and most likely contributing to the invasive phenotype. We suggest that MT1-MMP, together with TNF-α, should be investigated as potential therapeutic targets in AML.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...