Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Int J Mol Sci ; 23(2)2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35054772

RESUMO

Platelet factor 4 (CXCL4) is a chemokine abundantly stored in platelets. Upon injury and during atherosclerosis, CXCL4 is transported through the vessel wall where it modulates the function of vascular smooth muscle cells (VSMCs) by affecting proliferation, migration, gene expression and cytokine release. Variant CXCL4L1 is distinct from CXCL4 in function and expression pattern, despite a minor three-amino acid difference. Here, the effects of CXCL4 and CXCL4L1 on the phenotype and function of human VSMCs were compared in vitro. VSMCs were found to constitutively express CXCL4L1 and only exogenously added CXCL4 was internalized by VSMCs. Pre-treatment with heparin completely blocked CXCL4 uptake. A role of the putative CXCL4 receptors CXCR3 and DARC in endocytosis was excluded, but LDL receptor family members appeared to be involved in the uptake of CXCL4. Incubation of VSMCs with both CXCL4 and CXCL4L1 resulted in decreased expression of contractile marker genes and increased mRNA levels of KLF4 and NLRP3 transcription factors, yet only CXCL4 stimulated proliferation and calcification of VSMCs. In conclusion, CXCL4 and CXCL4L1 both modulate gene expression, yet only CXCL4 increases the division rate and formation of calcium-phosphate crystals in VSMCs. CXCL4 and CXCL4L1 may play distinct roles during vascular remodeling in which CXCL4 induces proliferation and calcification while endogenously expressed CXCL4L1 governs cellular homeostasis. The latter notion remains a subject for future investigation.


Assuntos
Calcinose , Proliferação de Células , Contração Muscular , Músculo Liso Vascular/metabolismo , Fator Plaquetário 4/fisiologia , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel/genética , Músculo Liso Vascular/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Fator Plaquetário 4/metabolismo
2.
Respir Med ; 171: 106098, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32777683

RESUMO

Platelets are essential mediators of inflammation and thrombosis. Chronic obstructive pulmonary disease (COPD) is a heterogeneous multisystem disease, causing significant morbidity and mortality worldwide. Recent evidence suggests that the lung is an important organ for platelet biogenesis. Cigarette smoking has been shown to induce platelet aggregation and decrease the capacity of mitochondrial electron transport system in platelets. Preclinical and clinical studies have suggested that platelets may contribute to the development of COPD through the breakdown of lung elastin by platelet factor 4, platelet activation and formation of platelet aggregates, and modulation of hypoxia signaling pathways. Recent large population studies have produced encouraging results indicating a potential role for aspirin in preventing exacerbations and delaying disease progression in patients with COPD. This review summarizes the information about the lung as an organ for platelet production, pathophysiological functions of platelets and platelet mediators in the development of COPD, and the most updated evidence on the utility of aspirin in patients with COPD.


Assuntos
Aspirina/administração & dosagem , Plaquetas/fisiologia , Ativação Plaquetária , Inibidores da Agregação Plaquetária , Agregação Plaquetária , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/etiologia , Aspirina/farmacologia , Plaquetas/metabolismo , Progressão da Doença , Elastina/metabolismo , Transporte de Elétrons , Humanos , Inflamação , Pulmão/citologia , Pulmão/metabolismo , Megacariócitos , Mitocôndrias/metabolismo , Doenças Mitocondriais , Fator Plaquetário 4/fisiologia , Fumar/efeitos adversos , Trombose
3.
Cancer Lett ; 491: 78-86, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-32726613

RESUMO

Malignant pleural effusion (MPE) is defined as the presence of tumor cells in pleural fluid and it is a fatal complication of advanced lung adenocarcinoma (LAC). To understand the immune response to the tumor in MPE, we compared the concentration of immunomodulatory factors in MPE of LAC and pleural effusion of heart failure (HF) patients by ELISA, and the proliferation and cytotoxic phenotype of T cells stimulated in the presence of LAC and HF pleural fluids by cytometry. Platelet factor 4 (PF4), vascular endothelial growth factor (VEGF), transforming growth factor beta (TGF-ß) and P-selectin levels were higher in LAC than in HF pleural fluids. However, plasmatic PF4 and P-selectin levels were similar in LAC and HF. VEGF positively correlated with TGF-ß and sPD-L1 in LAC but not in HF pleural fluids. LAC pleural fluids also inhibited T lymphocyte proliferation and cytotoxicity and reduced IL-17 production. PF4 levels inversely correlated with T cell function. The high content of PF4 in MPE was associated with poor prognosis. Our findings suggest that an impaired response of T lymphocytes induced by PF4 provides a significant advantage for tumor progression.


Assuntos
Adenocarcinoma de Pulmão/complicações , Neoplasias Pulmonares/complicações , Fator Plaquetário 4/fisiologia , Derrame Pleural Maligno/imunologia , Linfócitos T/imunologia , Adenocarcinoma de Pulmão/mortalidade , Idoso , Idoso de 80 Anos ou mais , Feminino , Insuficiência Cardíaca/imunologia , Humanos , Neoplasias Pulmonares/mortalidade , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Fator Plaquetário 4/análise , Derrame Pleural Maligno/mortalidade , Fator de Crescimento Transformador beta/análise , Fator A de Crescimento do Endotélio Vascular/análise
4.
Am J Physiol Cell Physiol ; 318(3): C542-C554, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31913695

RESUMO

Chemokines are a family of soluble cytokines that act as chemoattractants to guide the migration of cells, in particular of immune cells. However, chemokines are also involved in cell proliferation, differentiation, and survival. Chemokines are associated with a variety of human diseases including chronic inflammation, immune dysfunction, cancer, and metastasis. This review discusses the expression of CC and CXC chemokines in the tumor microenvironment and their supportive and inhibitory roles in tumor progression, angiogenesis, metastasis, and tumor immunity. We also specially focus on the diverse roles of CXC chemokines (CXCL9-11, CXCL4 and its variant CXCL4L1) and their two chemokine receptor CXCR3 isoforms, CXCR3-A and CXCR3-B. These two distinct isoforms have divergent roles in tumors, either promoting (CXCR3-A) or inhibiting (CXCR3-B) tumor progression. Their effects are mediated not only directly in tumor cells but also indirectly via the regulation of angiogenesis and tumor immunity. A full comprehension of their mechanisms of action is critical to further validate these chemokines and their receptors as biomarkers or therapeutic targets in cancer.


Assuntos
Biomarcadores Tumorais/fisiologia , Quimiocina CXCL9/fisiologia , Fator Plaquetário 4/fisiologia , Receptores CXCR3/fisiologia , Microambiente Tumoral/fisiologia , Animais , Progressão da Doença , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia
5.
Leuk Res ; 48: 107-12, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26803701

RESUMO

Platelet factor-4 (CXCL4/PF-4) is a member of CXC-chemokine family produced by megakaryocytic lineage and stored in platelet α-granules. Platelet stimulation by aggregating agents such as thrombin and ADP leads to CXCL4 secretion. CXCL4 plays several roles in coagulation, angiogenesis control, immune system modulation and spread of cancer. Megakaryocytes (Mks) are associated with the vascular niche in the bone marrow (BM) and are located in vicinity of BM sinusoids. Mk-derived CXCL4 is involved in several hematopoietic processes, including inhibition of megakaryopoiesis and maintenance of hematopoietic stem cell (HSC) quiescence. The major aim of this review article was to evaluate the role of CXCL4 in hematological malignancies, promotion of HSC quiescence as well as BM niche cells.


Assuntos
Células-Tronco Hematopoéticas/citologia , Megacariócitos/fisiologia , Fator Plaquetário 4/fisiologia , Células da Medula Óssea , Ciclo Celular , Neoplasias Hematológicas/patologia , Humanos , Nicho de Células-Tronco
6.
J Clin Invest ; 125(5): 1857-72, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25822018

RESUMO

Myelodysplastic syndromes and chronic myelomonocytic leukemia (CMML) are characterized by mutations in genes encoding epigenetic modifiers and aberrant DNA methylation. DNA methyltransferase inhibitors (DMTis) are used to treat these disorders, but response is highly variable, with few means to predict which patients will benefit. Here, we examined baseline differences in mutations, DNA methylation, and gene expression in 40 CMML patients who were responsive or resistant to decitabine (DAC) in order to develop a molecular means of predicting response at diagnosis. While somatic mutations did not differentiate responders from nonresponders, we identified 167 differentially methylated regions (DMRs) of DNA at baseline that distinguished responders from nonresponders using next-generation sequencing. These DMRs were primarily localized to nonpromoter regions and overlapped with distal regulatory enhancers. Using the methylation profiles, we developed an epigenetic classifier that accurately predicted DAC response at the time of diagnosis. Transcriptional analysis revealed differences in gene expression at diagnosis between responders and nonresponders. In responders, the upregulated genes included those that are associated with the cell cycle, potentially contributing to effective DAC incorporation. Treatment with CXCL4 and CXCL7, which were overexpressed in nonresponders, blocked DAC effects in isolated normal CD34+ and primary CMML cells, suggesting that their upregulation contributes to primary DAC resistance.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Genes Neoplásicos , Leucemia Mielomonocítica Crônica/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Medula Óssea/patologia , Metilação de DNA/efeitos dos fármacos , Análise Mutacional de DNA , DNA Intergênico/genética , Decitabina , Elementos Facilitadores Genéticos/genética , Feminino , Humanos , Leucemia Mielomonocítica Crônica/genética , Leucemia Mielomonocítica Crônica/metabolismo , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Fator Plaquetário 4/biossíntese , Fator Plaquetário 4/genética , Fator Plaquetário 4/fisiologia , Resultado do Tratamento , beta-Tromboglobulina/biossíntese , beta-Tromboglobulina/genética , beta-Tromboglobulina/fisiologia
7.
Invest Ophthalmol Vis Sci ; 56(3): 1956-64, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25711636

RESUMO

PURPOSE: To investigate the expression of platelet factor-4 variant (PF-4var/CXCL4L1) in epiretinal membranes from patients with proliferative diabetic retinopathy (PDR) and the role of PF-4var/CXCL4L1 in the regulation of blood-retinal barrier (BRB) breakdown in diabetic rat retinas and human retinal microvascular endothelial cells (HRMEC). METHODS: Rats were treated intravitreally with PF-4var/CXCL4L1 or the anti-vascular endothelial growth factor (VEGF) agent bevacizumab on the first day after diabetes induction. Blood-retinal barrier breakdown was assessed in vivo with fluorescein isothiocyanate (FITC)-conjugated dextran and in vitro in HRMEC by transendothelial electrical resistance and FITC-conjugated dextran cell permeability assay. Occludin, vascular endothelial (VE)-cadherin, hypoxia-inducible factor (HIF)-1α, VEGF, tumor necrosis factor (TNF)-α, receptor for advanced glycation end products (RAGE), caspase-3 levels, and generation of reactive oxygen species (ROS) were assessed by Western blot, enzyme-linked immunosorbent assays, or spectrophotometry. RESULTS: In epiretinal membranes, vascular endothelial cells and stromal cells expressed PF-4var/CXCL4L1. In vitro, HRMEC produced PF-4var/CXCL4L1 after stimulation with a combination of interleukin (IL)-1ß and TNF-α, and PF-4var/CXCL4L1 inhibited VEGF-mediated hyperpermeability in HRMEC. In rats, PF-4var/CXCL4L1 was as potent as bevacizumab in attenuating diabetes-induced BRB breakdown. This effect was associated with upregulation of occludin and VE-cadherin and downregulation of HIF-1α, VEGF, TNF-α, RAGE, and caspase-3, whereas ROS generation was not altered. CONCLUSIONS: Our findings suggest that increasing the intraocular PF-4var/CXCL4L1 levels early after the onset of diabetes protects against diabetes-induced BRB breakdown.


Assuntos
Barreira Hematorretiniana/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Retinopatia Diabética/tratamento farmacológico , Membrana Epirretiniana/metabolismo , Fator Plaquetário 4/uso terapêutico , Animais , Biomarcadores/metabolismo , Barreira Hematorretiniana/fisiologia , Caspase 3/metabolismo , Células Cultivadas , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , Humanos , Masculino , Fator Plaquetário 4/metabolismo , Fator Plaquetário 4/fisiologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
Cytokine Growth Factor Rev ; 26(1): 67-74, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25466649

RESUMO

Chronic liver allograft dysfunction is the leading cause of patient morbidity and late allograft loss after liver transplantation. The pathogenesis of chronic liver allograft dysfunction remains unknown. Recent studies have demonstrated that CXCL4 and its variant CXCL4L1 are involved in organ damage induced through inflammatory and immune responses throughout all stages of liver transplantation. CXCL4 and CXCL4L1 are low-molecular-weight proteins that have been implicated in hematopoiesis, angiostasis, organ fibrogenesis, mitogenesis, tumor growth and metastasis. The purpose of this review is to discuss the current status and future developments of research into the roles of CXCL4 and CXCL4L1 in the pathogenesis of chronic liver allograft dysfunction. The potential utilization of CXCL4 and CXCL4L1 as therapeutic targets for chronic liver allograft dysfunction will also be discussed.


Assuntos
Hepatopatias/fisiopatologia , Hepatopatias/terapia , Transplante de Fígado , Fator Plaquetário 4/fisiologia , Aloenxertos , Humanos , Hepatopatias/etiologia , Transplante de Fígado/efeitos adversos , Terapia de Alvo Molecular , Transdução de Sinais
9.
J Clin Invest ; 124(9): 3757-66, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25061876

RESUMO

The proteasome inhibiter bortezomib has been successfully used to treat patients with relapsed multiple myeloma; however, many of these patients become thrombocytopenic, and it is not clear how the proteasome influences platelet production. Here we determined that pharmacologic inhibition of proteasome activity blocks proplatelet formation in human and mouse megakaryocytes. We also found that megakaryocytes isolated from mice deficient for PSMC1, an essential subunit of the 26S proteasome, fail to produce proplatelets. Consistent with decreased proplatelet formation, mice lacking PSMC1 in platelets (Psmc1(fl/fl) Pf4-Cre mice) exhibited severe thrombocytopenia and died shortly after birth. The failure to produce proplatelets in proteasome-inhibited megakaryocytes was due to upregulation and hyperactivation of the small GTPase, RhoA, rather than NF-κB, as has been previously suggested. Inhibition of RhoA or its downstream target, Rho-associated protein kinase (ROCK), restored megakaryocyte proplatelet formation in the setting of proteasome inhibition in vitro. Similarly, fasudil, a ROCK inhibitor used clinically to treat cerebral vasospasm, restored platelet counts in adult mice that were made thrombocytopenic by tamoxifen-induced suppression of proteasome activity in megakaryocytes and platelets (Psmc1(fl/fl) Pdgf-Cre-ER mice). These results indicate that proteasome function is critical for thrombopoiesis, and suggest inhibition of RhoA signaling as a potential strategy to treat thrombocytopenia in bortezomib-treated multiple myeloma patients.


Assuntos
Complexo de Endopeptidases do Proteassoma/fisiologia , Trombopoese , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Fator Plaquetário 4/fisiologia , Glicoproteína IIb da Membrana de Plaquetas/fisiologia , Inibidores de Proteassoma/farmacologia , Trombopoese/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP
10.
Arterioscler Thromb Vasc Biol ; 34(3): 571-80, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371083

RESUMO

OBJECTIVE: A growing body of evidence indicates that platelets contribute to the onset and progression of atherosclerosis by modulating immune responses. We aimed to elucidate the effects of oxidized low-density lipoprotein (OxLDL) on platelet-monocyte interactions and the consequences of these interactions on platelet phagocytosis, chemokine release, monocyte extravasation, and foam cell formation. APPROACH AND RESULTS: Confocal microscopy and flow cytometric analysis revealed that in vitro and in vivo stimulation with OxLDL resulted in rapid formation of platelet-monocyte aggregates, with a preference for CD16+ monocyte subsets. This platelet-monocyte interaction facilitated OxLDL uptake by monocytes, in a process that involved platelet CD36-OxLDL interaction, release of chemokines, such as CXC motif ligand 4, direct platelet-monocyte interaction, and phagocytosis of platelets. Inhibition of cyclooxygenase with acetylsalicylic acid and antagonists of ADP receptors, P2Y1 and P2Y12, partly abrogated OxLDL-induced platelet-monocyte aggregates and platelet-mediated lipid uptake in monocytes. Platelets also enhanced OxLDL-induced monocyte transmigration across an endothelial monolayer via direct interaction with monocytes in a transwell assay. Importantly, in LDLR(-/-) mice, platelet depletion resulted in a significant decrease of peritoneal macrophage recruitment and foam cell formation in a thioglycollate-elicited peritonitis model. In platelet-depleted wild-type mice, transfusion of ex vivo OxLDL-stimulated platelets induced monocyte extravasation to a higher extent when compared with resting platelets. CONCLUSIONS: Our results on OxLDL-mediated platelet-monocyte aggregate formation, which promoted phenotypic changes in monocytes, monocyte extravasation and enhanced foam cell formation in vitro and in vivo, provide a novel mechanism for how platelets potentiate key steps of atherosclerotic plaque development and plaque destabilization.


Assuntos
Plaquetas/fisiologia , Quimiotaxia/efeitos dos fármacos , Células Espumosas/efeitos dos fármacos , Lipoproteínas LDL/farmacologia , Monócitos/efeitos dos fármacos , Animais , Aspirina/farmacologia , Aterosclerose/fisiopatologia , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Antígeno CD11b/fisiologia , Células Cultivadas , Quimiocinas/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Células Espumosas/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipoproteínas LDL/toxicidade , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/citologia , Selectina-P/sangue , Peritonite/induzido quimicamente , Peritonite/patologia , Fagocitose/efeitos dos fármacos , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Fator Plaquetário 4/fisiologia , Transfusão de Plaquetas , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores de LDL/deficiência , Receptores de LDL/genética , Migração Transendotelial e Transepitelial/efeitos dos fármacos
11.
J Biol Chem ; 288(19): 13522-33, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23536183

RESUMO

BACKGROUND: CXCL4L1 is a highly potent anti-angiogenic and anti-tumor chemokine, and its structural information is unknown. RESULTS: CXCL4L1 x-ray structure is determined, and it reveals a previously unrecognized chemokine structure adopting a novel C-terminal helix conformation. CONCLUSION: The alternative helix conformation enhances the anti-angiogenic activity of CXCL4L1 by reducing the glycosaminoglycan binding ability. SIGNIFICANCE: Chemokine C-terminal helix orientation is critical in regulating their functions. Chemokines, a subfamily of cytokines, are small, secreted proteins that mediate a variety of biological processes. Various chemokines adopt remarkable conserved tertiary structure comprising an anti-parallel ß-sheet core domain followed by a C-terminal helix that packs onto the ß-sheet. The conserved structural feature has been considered critical for chemokine function, including binding to cell surface receptor. The recently isolated variant, CXCL4L1, is a homologue of CXCL4 chemokine (or platelet factor 4) with potent anti-angiogenic activity and differed only in three amino acid residues of P58L, K66E, and L67H. In this study we show by x-ray structural determination that CXCL4L1 adopts a previously unrecognized structure at its C terminus. The orientation of the C-terminal helix protrudes into the aqueous space to expose the entire helix. The alternative helix orientation modifies the overall chemokine shape and surface properties. The L67H mutation is mainly responsible for the swing-out effect of the helix, whereas mutations of P58L and K66E only act secondarily. This is the first observation that reports an open conformation of the C-terminal helix in a chemokine. This change leads to a decrease of its glycosaminoglycan binding properties and to an enhancement of its anti-angiogenic and anti-tumor effects. This unique structure is recent in evolution and has allowed CXCL4L1 to gain novel functional properties.


Assuntos
Fator Plaquetário 4/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas Angiogênicas/química , Cristalografia por Raios X , Cistina/química , Ditiotreitol/química , Heparina/química , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oxirredução , Fator Plaquetário 4/genética , Fator Plaquetário 4/fisiologia , Ligação Proteica , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Substâncias Redutoras/química
12.
PLoS One ; 7(12): e51361, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23300543

RESUMO

The development of a megakaryocyte lineage specific Cre deleter, using the Pf4 (CXCL4) promoter (Pf4-Cre), was a significant step forward in the specific analysis of platelet and megakaryocyte cell biology. However, in the present study we have employed a sensitive reporter-based approach to demonstrate that Pf4-Cre also recombines in a significant proportion of both fetal liver and bone marrow hematopoietic stem cells (HSCs), including the most primitive fraction containing the long-term repopulating HSCs. Consequently, we demonstrate that Pf4-Cre activity is not megakaryocyte lineage-specific but extends to other myeloid and lymphoid lineages at significant levels between 15-60%. Finally, we show for the first time that Pf4 transcripts are present in adult HSCs and primitive hematopoietic progenitor cells. These results have fundamental implications for the use of the Pf4-Cre mouse model and for our understanding of a possible role for Pf4 in the development of the hematopoietic lineage.


Assuntos
Células da Medula Óssea/citologia , Linhagem da Célula , Feto/citologia , Células-Tronco Hematopoéticas/citologia , Integrases/metabolismo , Fígado/citologia , Megacariócitos/citologia , Fator Plaquetário 4/fisiologia , Animais , Plaquetas/metabolismo , Células da Medula Óssea/metabolismo , Células Cultivadas , DNA/genética , Feto/metabolismo , Citometria de Fluxo , Células-Tronco Hematopoéticas/metabolismo , Fígado/metabolismo , Linfócitos/citologia , Linfócitos/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
13.
Int J Radiat Oncol Biol Phys ; 80(5): 1533-40, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21740995

RESUMO

PURPOSE: Factors affecting the severity of radiation-induced thrombocytopenia (RIT) are not well described. We address whether platelet factor 4 (PF4; a negative paracrine for megakaryopoiesis) affects platelet recovery postradiation. METHODS AND MATERIALS: Using conditioned media from irradiated bone marrow (BM) cells from transgenic mice overexpressing human (h) PF4 (hPF4+), megakaryocyte colony formation was assessed in the presence of this conditioned media and PF4 blocking agents. In a model of radiation-induced thrombocytopenia, irradiated mice with varying PF4 expression levels were treated with anti-hPF4 and/or thrombopoietin (TPO), and platelet count recovery and survival were examined. RESULTS: Conditioned media from irradiated BM from hPF4+ mice inhibited megakaryocyte colony formation, suggesting that PF4 is a negative paracrine released in RIT. Blocking with an anti-hPF4 antibody restored colony formation of BM grown in the presence of hPF4+ irradiated media, as did antibodies that block the megakaryocyte receptor for PF4, low-density lipoprotein receptor-related protein 1 (LRP1). Irradiated PF4 knockout mice had higher nadir platelet counts than irradiated hPF4+/knockout litter mates (651 vs. 328 × 106/mcL, p = 0.02) and recovered earlier (15 days vs. 22 days, respectively, p <0.02). When irradiated hPF4+ mice were treated with anti-hPF4 antibody and/or TPO, they showed less severe thrombocytopenia than untreated mice, with improved survival and time to platelet recovery, but no additive effect was seen. CONCLUSIONS: Our studies show that in RIT, damaged megakaryocytes release PF4 locally, inhibiting platelet recovery. Blocking PF4 enhances recovery while released PF4 from megakaryocytes limits TPO efficacy, potentially because of increased release of PF4 stimulated by TPO. The clinical value of blocking this negative paracrine pathway post-RIT remains to be determined.


Assuntos
Plaquetas/efeitos da radiação , Megacariócitos/efeitos da radiação , Fator Plaquetário 4/fisiologia , Trombocitopenia/etiologia , Animais , Plaquetas/citologia , Medula Óssea/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias/métodos , Meios de Cultivo Condicionados/efeitos da radiação , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contagem de Plaquetas , Fator Plaquetário 4/antagonistas & inibidores , Receptores de LDL/antagonistas & inibidores , Trombocitopenia/sangue , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Trombopoese/fisiologia , Trombopoese/efeitos da radiação , Trombopoetina/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores
14.
Cytokine Growth Factor Rev ; 22(1): 1-18, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21111666

RESUMO

Chemokines are chemotactic cytokines which recruit leukocytes to inflammatory sites. They also affect tumor development and metastasis by acting as growth factor, by attracting pro- or anti-tumoral leukocytes or by influencing angiogenesis. Platelet factor-4 (CXCL4/PF-4) was the first chemokine shown to inhibit angiogenesis. CXCL4L1/PF-4var, recently isolated from thrombin-stimulated platelets, differing from authentic CXCL4/PF-4 in three carboxy-terminally located amino acids, was found to be more potent than CXCL4/PF-4 in inhibiting angiogenesis and tumor growth. Both glycosaminoglycans (GAG) and CXCR3 are implicated in the activities of the PF-4 variants. This report reviews the current knowledge on the role of CXCL4/PF-4 and CXCL4L1/PF-4var in physiological and pathological processes. In particular, the role of CXCL4/PF-4 in cancer, heparin-induced thrombocytopenia and atherosclerosis is described.


Assuntos
Inflamação/etiologia , Neoplasias/etiologia , Neovascularização Patológica/etiologia , Neovascularização Fisiológica/genética , Fator Plaquetário 4/fisiologia , Animais , Humanos , Inflamação/complicações , Inflamação/genética , Modelos Biológicos , Neoplasias/irrigação sanguínea , Neoplasias/complicações , Neoplasias/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/terapia , Neovascularização Fisiológica/fisiologia , Fator Plaquetário 4/genética , Fator Plaquetário 4/farmacologia , Fator Plaquetário 4/uso terapêutico
15.
J Immunol ; 184(9): 4810-8, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20335529

RESUMO

In atherosclerotic arteries, blood monocytes differentiate to macrophages in the presence of growth factors, such as macrophage colony-stimulation factor (M-CSF), and chemokines, such as platelet factor 4 (CXCL4). To compare the gene expression signature of CXCL4-induced macrophages with M-CSF-induced macrophages or macrophages polarized with IFN-gamma/LPS (M1) or IL-4 (M2), we cultured primary human peripheral blood monocytes for 6 d. mRNA expression was measured by Affymetrix gene chips, and differences were analyzed by local pooled error test, profile of complex functionality, and gene set enrichment analysis. Three hundred seventy-five genes were differentially expressed between M-CSF- and CXCL4-induced macrophages; 206 of them overexpressed in CXCL4 macrophages coding for genes implicated in the inflammatory/immune response, Ag processing and presentation, and lipid metabolism. CXCL4-induced macrophages overexpressed some M1 and M2 genes and the corresponding cytokines at the protein level; however, their transcriptome clustered with neither M1 nor M2 transcriptomes. They almost completely lost the ability to phagocytose zymosan beads. Genes linked to atherosclerosis were not consistently upregulated or downregulated. Scavenger receptors showed lower and cholesterol efflux transporters showed higher expression in CXCL4- than M-CSF-induced macrophages, resulting in lower low-density lipoprotein content. We conclude that CXCL4 induces a unique macrophage transcriptome distinct from known macrophage types, defining a new macrophage differentiation that we propose to call M4.


Assuntos
Diferenciação Celular/imunologia , Perfilação da Expressão Gênica , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Fator Plaquetário 4/fisiologia , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/patologia , Biomarcadores/sangue , Diferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Polaridade Celular/genética , Polaridade Celular/imunologia , Células Cultivadas , Progressão da Doença , Humanos , Fator Estimulador de Colônias de Macrófagos/sangue , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/citologia , Monócitos/citologia , Fator Plaquetário 4/sangue
16.
PLoS One ; 3(7): e2657, 2008 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-18648521

RESUMO

The human CXC-chemokine CXCL4 is a potent inhibitor of tumor-induced angiogenesis. Considering that CXCL4 is sequestered in platelet alpha-granules and released following platelet activation in the vicinity of vessel wall injury, we tested the hypothesis that CXCL4 might function as a ligand for integrins. Integrins are a family of adhesion receptors that play a crucial role in angiogenesis by regulating early angiogenic processes, such as endothelial cell adhesion and migration. Here, we show that CXCL4 interacts with alphavbeta3 on the surface of alphavbeta3-CHO. More importantly, human umbilical vein endothelial cells adhere to immobilized CXCL4 through alphavbeta3 integrin, and also through other integrins, such as alphavbeta5 and alpha5beta1. We further demonstrate that CXCL4-integrin interaction is of functional significance in vitro, since immobilized CXCL4 supported endothelial cell spreading and migration in an integrin-dependent manner. Soluble CXCL4, in turn, inhibits integrin-dependent endothelial cell adhesion and migration. As a whole, our study identifies integrins as novel receptors for CXCL4 that may contribute to its antiangiogenic effect.


Assuntos
Quimiocinas CXC/fisiologia , Integrinas/metabolismo , Neovascularização Patológica , Fator Plaquetário 4/fisiologia , Animais , Células CHO , Adesão Celular , Movimento Celular , Quimiocinas CXC/metabolismo , Cricetinae , Cricetulus , Endotélio Vascular/metabolismo , Humanos , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Fator Plaquetário 4/metabolismo , Receptores de Vitronectina/metabolismo
17.
Nihon Rinsho ; 65(12): 2195-201, 2007 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-18069260

RESUMO

Accumulating evidences suggest that many molecules are working as inhibitors of proliferation in myeloma cells e.g., PTEN, mTOR(PI3-kinase signal molecules), p53, RB1, INK4 family and KIP/CIP family (cell cycle check point molecules), PF4 (inhibitor of angiogenesis). In this review, significance of these molecules in myeloma is summarized. Additionally, our finding of growth inhibitory effect by PU.1 is explained.


Assuntos
Divisão Celular/genética , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Plasmócitos/citologia , Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Humanos , PTEN Fosfo-Hidrolase/fisiologia , Plasmócitos/patologia , Fator Plaquetário 4/fisiologia , Proteínas Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Serina-Treonina Quinases TOR , Transativadores/fisiologia , Proteína Supressora de Tumor p53/fisiologia
18.
Thromb Haemost ; 97(5): 704-13, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17479180

RESUMO

Undoubtedly, platelets are key elements in the regulation of thrombosis and haemostasis. Along with their primary task to prevent blood loss from injured vessels, platelets have emerged as regulators of a variety of processes in the vasculature. Multiple challenges, from the contact and adhesion to subendothelial matrix after injury of the vessel wall, to interactions with blood cells in inflammatory conditions, result in platelet activation with concomitant shape change and release of numerous substances. Among these, chemokines have been found to modulate several processes in the vasculature, such as atherosclerosis and angiogenesis. In particular, the chemokines connective tissue activating protein III (CTAP-III) and its precursors, or truncation products (CXCL7), platelet factor 4, (PF4, CXCL4) and its variant PF4alt (CXCL4L1) or regulated upon activation and normal T cell expressed and secreted (RANTES, CCL5), have been investigated thoroughly. Defined common properties as their aptitude to bind glycosaminoglycans or their predisposition to associate and form homooligomers are pre-requisites for their role in the vasculature and function in vivo. The current review summarizes the development of these single chemokines, and their cooperative effects that may in part be dependent on their physical interactions.


Assuntos
Plaquetas/imunologia , Quimiocinas/sangue , Animais , Aterosclerose/etiologia , Aterosclerose/imunologia , Humanos , Camundongos , Modelos Imunológicos , Neovascularização Fisiológica , Ativação Plaquetária , Fator Plaquetário 4/fisiologia , Receptores de Quimiocinas/sangue , Transdução de Sinais
19.
Blood ; 110(4): 1153-60, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17495129

RESUMO

Platelet factor 4 (PF4) is a negative regulator of megakaryopoiesis in vitro. We have now examined whether PF4 regulates megakaryopoiesis in vivo by studying PF4 knockout mice and transgenic mice that overexpress human (h) PF4. Steady-state platelet count and thrombocrit in these animals was inversely related to platelet PF4 content. Growth of megakaryocyte colonies was also inversely related to platelet PF4 content. Function-blocking anti-PF4 antibody reversed this inhibition of megakaryocyte colony growth, indicating the importance of local PF4 released from developing megakaryocytes. The effect of megakaryocyte damage and release of PF4 on 5-fluorouracil-induced marrow failure was then examined. Severity of thrombocytopenia and time to recovery of platelet counts were inversely related to initial PF4 content. Recovery was faster and more extensive, especially in PF4-overexpressing mice, after treatment with anti-PF4 blocking antibodies, suggesting a means to limit the duration of such a chemotherapy-induced thrombocytopenia, especially in individuals with high endogenous levels of PF4. We found that approximately 8% of 250 healthy adults have elevated (> 2 times average) platelet PF4 content. These individuals with high levels of platelet PF4 may be especially sensitive to developing thrombocytopenia after bone marrow injury and may benefit from approaches that block the effects of released PF4.


Assuntos
Comunicação Autócrina , Hematopoese/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Fator Plaquetário 4/farmacologia , Adulto , Animais , Células Cultivadas , Humanos , Masculino , Megacariócitos/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contagem de Plaquetas , Fator Plaquetário 4/imunologia , Fator Plaquetário 4/fisiologia , Trombopoetina/sangue
20.
J Biol Chem ; 282(1): 710-20, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17090548

RESUMO

Platelet factor-4 (PF4/CXCL4) is an orphan chemokine released in large quantities in the vicinity of growing blood clots. Coagulation of plasma supplemented with a matching amount of PF4 results in a translucent jelly-like clot. Saturating amounts of PF4 reduce the porosity of the fibrin network 4.4-fold and decrease the values of the elastic and loss moduli by 31- and 59-fold, respectively. PF4 alters neither the cleavage of fibrinogen by thrombin nor the cross-linking of protofibrils by activated factor XIII but binds to fibrin and dramatically transforms the structure of the ensuing network. Scanning electron microscopy showed that PF4 gives rise to a previously unreported pattern of polymerization where fibrin assembles to form a sealed network. The subunits constituting PF4 form a tetrahedron having at its corners a RPRH motif that mimics (in reverse orientation) the Gly-His-Arg-Pro-amide peptides that co-crystallize with fibrin. Molecular modeling showed that PF4 could be docked to fibrin with remarkable complementarities and absence of steric clashes, allowing the assembly of irregular polymers. Consistent with this hypothesis, as little as 50 microm the QVRPRHIT peptide derived from PF4 affects the polymerization of fibrin.


Assuntos
Coagulação Sanguínea , Fibrina/química , Fator Plaquetário 4/fisiologia , Reagentes de Ligações Cruzadas/farmacologia , Dimerização , Fibrinogênio/química , Raios gama , Humanos , Cinética , Microscopia Eletrônica de Varredura , Modelos Moleculares , Peptídeos/química , Fator Plaquetário 4/química , Polímeros/química , Ligação Proteica , Eletricidade Estática
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA