Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Adv Exp Med Biol ; 1408: 49-63, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093421

RESUMO

Catecholamine stimulation over adrenergic receptors results in a state of hypercoagulability. Chronic stress involves the release and increase in circulation of catecholamines and other stress related hormones. Numerous observational studies in human have related stressful scenarios to several coagulation variables, but controlled stimulation with agonists or antagonists to adrenergic receptors are scarce. This systematic review is aimed at presenting an updated appraisal of the effect of adrenergic receptor modulation on variables related to human hemostasis by systematically reviewing the effect of adrenergic receptor-targeting drugs on scale variables related to hemostasis. By searching 3 databases for articles published between January 1st 2011 and February 16th, 2022 reporting effects on coagulation parameters from stimulation with α- or ß-adrenergic receptor targeting drugs in humans regardless of baseline condition, excluding records different from original research and those not addressing the main aim of this systematic review. Risk of bias assessed using the Revised Cochrane risk-of-bias tool for randomized trials (RoB 2). Tables describing a pro-thrombotic anti-fibrinolytic state induced after ß-adrenergic receptor agonist stimulation and the opposite after α1-, ß-adrenergic receptor antagonist stimulation were synthesized from 4 eligible records by comparing hemostasis-related variables to their baseline. Notwithstanding this low number of records, experimental interventions included were sound and mostly unbiased, results were coherent, and outcomes were biologically plausible. In summary, this systematic review provides a critical systematic assessment and an updated elaboration, and its shortcomings highlight the need for further investigation in the field of hematology.


Assuntos
Adrenérgicos , Hemostasia , Receptores Adrenérgicos , Catecolaminas , Receptores Adrenérgicos/metabolismo , Adrenérgicos/uso terapêutico , Hemostasia/efeitos dos fármacos , Humanos , Estresse Fisiológico , Coagulação Sanguínea
2.
Adv Exp Med Biol ; 1408: 235-249, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093431

RESUMO

The organization of a circadian system includes an endogenous pacemaker system, input pathways for environmental synchronizing (entraining) stimuli, and output pathways through which the clock regulates physiological and behavioral processes, for example, the glucose-sensing mechanism in the liver. The liver is the central regulator of metabolism and one of our peripherals clocks. In mammals, central to this pacemaker are the transcription factors Circadian Locomotor Output Cycles Kaput (CLOCK) and BMAL1 (Brain and Muscle ARNT-Like 1). BMAL1 dimerizes with CLOCK, and this heterodimer then binds to the E-box promoter elements (CACGTG) present in clock and clock-controlled genes (CCGs). However, we are just beginning to understand how output pathways and regulatory mechanisms of CCGs are involved in rhythmic physiological processes. Glucokinase (GCK) is a fundamental enzyme in glucose homeostasis, catalyzing the high Km phosphorylation of glucose and allowing its storage. Moreover, gck is a dependent circadian gene. This study aims to determine the contribution of clock genes to hepatic gck expression and to define the specific role of E-box sequences on the circadian regulation of hepatic gck. Results showed that gck expression follows a circadian rhythm in rat hepatocytes in vitro. Accordingly, bmal1 expression induces the glucokinase circadian rhythmic expression in hepatocytes and the analysis of human and rat gck promoters, indicating the presence of E-box regions. Moreover, the basal activity of gck promoter was increased by clock/bmal1 co-transfection but inhibited by Period1/Period2 (per1/per2) co-transfection. Thus, the data suggest that the clock proteins tightly regulate the transcriptional activity of the gck promoter.


Assuntos
Fatores de Transcrição ARNTL , Elementos E-Box , Ratos , Humanos , Animais , Fatores de Transcrição ARNTL/genética , Glucoquinase , Ritmo Circadiano/fisiologia , Glucose , Regulação da Expressão Gênica , Mamíferos/genética
3.
Adv Exp Med Biol ; 1408: 309-328, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093435

RESUMO

Transforming growth factor-beta1 (TGF-ß) regulates a plethora of cell-intrinsic processes that modulate tumor progression in a context-dependent manner. Thus, although TGF-ß acts as a tumor suppressor in the early stages of tumorigenesis, in late stages, this factor promotes tumor progression and metastasis. In addition, TGF-ß also impinges on the tumor microenvironment by modulating the immune system. In this aspect, TGF-ß exhibits a potent immunosuppressive effect, which allows both cancer cells to escape from immune surveillance and confers resistance to immunotherapy. While TGF-ß inhibits the activation and antitumoral functions of T-cell lymphocytes, dendritic cells, and natural killer cells, it promotes the generation of T-regulatory cells and myeloid-derived suppressor cells, which hinder antitumoral T-cell activities. Moreover, TGF-ß promotes tumor-associated macrophages and neutrophils polarization from M1 into M2 and N1 to N2, respectively. Altogether, these effects contribute to the generation of an immunosuppressive tumor microenvironment and support tumor promotion. This review aims to analyze the relevant evidence on the complex role of TGF-ß in cancer immunology, the current outcomes of combined immunotherapies, and the anti-TGF-ß therapies that may improve the success of current and new oncotherapies.


Assuntos
Linfócitos T Reguladores , Fator de Crescimento Transformador beta1 , Humanos , Células Matadoras Naturais , Carcinogênese , Imunoterapia , Fator de Crescimento Transformador beta/fisiologia , Microambiente Tumoral
4.
Adv Exp Med Biol ; 1408: 163-181, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093427

RESUMO

Dexmedetomidine is an adrenergic receptor agonist that has been regarded as neuroprotective in several studies without an objective measure to it. Thus, the aim of this meta-analysis was to analyze and quantify the current evidence for the neuroprotective effects of dexmedetomidine in animals. The search was performed by querying the National Library of Medicine. Studies were included based on their language, significancy of their results, and complete availability of data on animal characteristics and interventions. Risk of bias was assessed using SYRCLE's risk of bias tool and certainty was assessed using the ARRIVE Guidelines 2.0. Synthesis was performed by calculating pooled standardized mean difference and presented in forest plots and tables. The number of eligible records included per outcome is the following: 22 for IL-1ß, 13 for IL-6, 19 for apoptosis, 7 for oxidative stress, 7 for Escape Latency, and 4 for Platform Crossings. At the cellular level, dexmedetomidine was found protective against production of IL-1ß (standardized mean difference (SMD) = - 4.3 [- 4.8; - 3.7]) and IL-6 (SMD = - 5.6 [- 6.7; - 4.6]), apoptosis (measured through TUNEL, SMD = - 6.0 [- 6.8; - 4.6]), and oxidative stress (measured as MDA production, SMD = - 2.0 [- 2.4; - 1.4]) exclusively in the central nervous system. At the organism level, dexmedetomidine improved behavioral outcomes measuring escape latency (SMD = - 2.4 [- 3.3; - 1.6]) and number of platform crossings (SMD = 9.1 [- 6.8; - 11.5]). No eligible study had high risk of bias and certainty was satisfactory for reproducibility in all cases. This meta-analysis highlights the complexity of adrenergic stimulation and sheds light into the mechanisms potentiated by dexmedetomidine, which could be exploited for improving current neuroprotective formulations.


Assuntos
Dexmedetomidina , Fármacos Neuroprotetores , Estados Unidos , Interleucina-6 , Reprodutibilidade dos Testes
5.
Adv Exp Med Biol ; 1408: 273-290, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37093433

RESUMO

The Philadelphia-negative myeloproliferative neoplasms (MPNs), defined as clonal disorders of the hematopoietic stem cells, are characterized by the proliferation of mature myeloid cells in the bone marrow and a chronic inflammatory status impacting the initiation, progression, and symptomatology of the malignancies. There are three main entities defined as essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), and genetically classified by JAK2V617F, CALR, or MPL mutations. In MPNs, due to the overproduction of inflammatory cytokines by the neoplastic cells and non-transformed immune cells, chronic inflammation may provoke the generation and expansion of myeloid-derived suppressors cells (MDSCs) that highly influence the adaptive immune response. Although peripheral blood MDSC levels are elevated, their frequency in the bone marrow of MPNs patients is not well elucidated yet. Our results indicated increased levels of total (T)-MDSCs (CD33+HLA-DR-/low) and polymorphonuclear (PMN)-MDSCs (CD33+/HLA-DRlow/CD15+/CD14-) in the bone marrow and peripheral blood of all three types of MPNs malignancies. However, these bone marrow MDSCs-increased frequencies did not correlate with the clinical parameters, such as hepatomegaly, leukocytes, hemoglobin, or platelet levels, or with JAK2 and CALR mutations. Besides, bone marrow MDSCs, from ET, PV, and PMF patients, exhibited immunosuppressive function, determined as T-cell proliferation inhibition. Notably, the highest T-MDSCs and PMN-MDSC levels were found in PMF samples, and the increased MDSCs frequency strongly correlated with the degree of myelofibrosis. Thus, these data together indicate that the immunosuppressive MDSCs population is increased in the bone marrow of MPNs patients and may be implicated in generating a fibrotic microenvironment.


Assuntos
Células Supressoras Mieloides , Transtornos Mieloproliferativos , Policitemia Vera , Mielofibrose Primária , Trombocitemia Essencial , Humanos , Mielofibrose Primária/genética , Mielofibrose Primária/patologia , Medula Óssea/patologia , Células Supressoras Mieloides/patologia , Transtornos Mieloproliferativos/genética , Policitemia Vera/genética , Policitemia Vera/patologia , Trombocitemia Essencial/genética , Trombocitemia Essencial/patologia , Janus Quinase 2/genética , Mutação , Microambiente Tumoral
6.
Dev Dyn ; 251(1): 105-124, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33797140

RESUMO

Transforming growth factor-beta1 (TGF-ß1) plays a crucial role in tumor progression. It can inhibit early cancer stages but promotes tumor growth and development at the late stages of tumorigenesis. TGF-ß1 has a potent immunosuppressive function within the tumor microenvironment that largely contributes to tumor cells' immune escape and reduction in cancer immunotherapy responses. Likewise, myeloid-derived suppressor cells (MDSCs) have been postulated as leading tumor promoters and a hallmark of cancer immune evasion mechanisms. This review attempts to analyze the prominent roles of both TGF-ß1 and MDSCs and their interplay in cancer immunity. Furthermore, therapies against either TGF-ß1 or MDSCs, and their potential synergistic combination with immunotherapies are discussed. Simultaneous TGF-ß1 and MDSCs inhibition suggest a potential improvement in immunotherapy or subverted tumor immune resistance.


Assuntos
Células Supressoras Mieloides , Neoplasias , Humanos , Células Supressoras Mieloides/patologia , Neoplasias/patologia , Neoplasias/terapia , Fator de Crescimento Transformador beta1 , Evasão Tumoral , Microambiente Tumoral
7.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-34206393

RESUMO

BACKGROUND: Chronic inflammation has been recognized in neoplastic disorders, including myeloproliferative neoplasm (MPN), as an important regulator of angiogenesis. AIMS: We investigated the influence of vascular endothelial growth factor (VEGF) and pro-inflammatory interleukin-6 (IL-6) on the expression of angiogenic factors, as well as inflammation-related signaling in mononuclear cells (MNC) of patients with MPN and JAK2V617F positive human erythroleukemic (HEL) cells. RESULTS: We found that IL-6 did not change the expression of angiogenic factors in the MNC of patients with MPN and HEL cells. However, IL-6 and the JAK1/2 inhibitor Ruxolitinib significantly increased angiogenic factors-endothelial nitric oxide synthase (eNOS), VEGF, and hypoxia-inducible factor-1 alpha (HIF-1α)-in patients with polycythemia vera (PV). Furthermore, VEGF significantly increased the expression of HIF-1α and eNOS genes, the latter inversely regulated by PI3K and mTOR signaling in the MNC of primary myelofibrosis (PMF). VEGF and inhibitors of inflammatory JAK1/2, PI3K, and mTOR signaling reduced the eNOS protein expression in HEL cells. VEGF also decreased the expression of eNOS and HIF-1α proteins in the MNC of PMF. In contrast, VEGF increased eNOS and HIF-1α protein expression in the MNC of patients with PV, which was mediated by the inflammatory signaling. VEGF increased the level of IL-6 immunopositive MNC of MPN. In summary, VEGF conversely regulated gene and protein expression of angiogenic factors in the MNC of PMF, while VEGF increased angiogenic factor expression in PV mediated by the inflammation-related signaling. CONCLUSION: The angiogenic VEGF induction of IL-6 supports chronic inflammation that, through positive feedback, further promotes angiogenesis with concomitant JAK1/2 inhibition.


Assuntos
Transtornos Mieloproliferativos/etiologia , Transtornos Mieloproliferativos/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Biomarcadores , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação , Transtornos Mieloproliferativos/patologia
8.
Cell Biochem Funct ; 38(4): 362-372, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31885098

RESUMO

This study has been performed to determine the mechanism of activation of the myeloid related S100A proteins by inflammatory cytokines in myeloproliferative neoplasm (MPN). Besides microarray analysis of MPN-derived CD34+ cells, we analysed the pro-inflammatory IL6 and anti-inflammatory IL10 dependence of NF-κB, PI3K-AKT, and JAK-STAT signalling during induction of S100A proteins in mononuclear cells of MPN, by immunoblotting and flow cytometry. We observed the reduced gene expression linked to NF-κB and inflammation signalling in MPN-derived CD34+ cells. Both IL6 and IL10 reduced S100A8 and 100A9 protein levels mediated via NF-κB and PI3K signalling, respectively, in mononuclear cells of essential thrombocythemia (ET). We also determined the increased percentage of S100A8 and S100A9 positive granulocytes in ET and primary myelofibrosis, upgraded by the JAK2V617F mutant allele burden. S100A8/9 heterodimer induced JAK1/2-dependent mitotic arrest of the ET-derived granulocytes. SIGNIFICANCE OF THE STUDY: We demonstrated that inflammation reduced the myeloid related S100A8/9 proteins by negative feedback mechanism in ET. S100A8/9 can be a diagnostic marker of inflammation in MPN, supported by the concomitant NF-κB and JAK1/2 signalling inhibition in regulation of myeloproliferation and therapy of MPN.


Assuntos
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Interleucina-6/metabolismo , Leucócitos Mononucleares/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Trombocitemia Essencial/metabolismo , Substituição de Aminoácidos , Calgranulina A/genética , Calgranulina B/genética , Feminino , Humanos , Interleucina-6/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Mutação de Sentido Incorreto , NF-kappa B/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Trombocitemia Essencial/genética , Trombocitemia Essencial/patologia
9.
Cancer Immunol Immunother ; 68(4): 645-660, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30003321

RESUMO

Mounting evidence has accumulated on the critical role of the different myeloid cells in the regulation of the cancerous process, and in particular in the modulation of the immune reaction to cancer. Myeloid cells are a major component of host cells infiltrating tumors, interacting with each other, with tumor cells and other stromal cells, and demonstrating a prominent plasticity. We describe here various myeloid regulatory cells (MRCs) in mice and human as well as their relevant therapeutic targets. We first address the role of the monocytes and macrophages that can contribute to angiogenesis, immunosuppression and metastatic dissemination. Next, we discuss the differential role of neutrophil subsets in tumor development, enhancing the dual and sometimes contradicting role of these cells. A heterogeneous population of immature myeloid cells, MDSCs, was shown to be generated and accumulated during tumor progression as well as to be an important player in cancer-related immune suppression. Lastly, we discuss the role of myeloid DCs, which can either contribute to effective anti-tumor responses or play a more regulatory role. We believe that MRCs play a critical role in cancer-related immune regulation and suggest that future anti-cancer therapies will focus on these abundant cells.


Assuntos
Comunicação Celular/imunologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Animais , Biomarcadores , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Neoplasias/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo
10.
Cell Biol Int ; 43(2): 192-206, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30571852

RESUMO

Myeloproliferative neoplasms (MPNs) are developing resistance to therapy by JAK1/2 inhibitor ruxolitinib. To explore the mechanism of ruxolitinib's limited effect, we examined the JAK1/2 mediated induction of proliferation related ERK1/2 and AKT signaling by proinflammatory interleukin-6 (IL-6) in MPN granulocytes and JAK2V617F mutated human erythroleukemia (HEL) cells. We found that JAK1/2 or JAK2 inhibition prevented the IL-6 activation of STAT3 and AKT pathways in polycythemia vera and HEL cells. Further, we showed that these inhibitors also blocked the IL-6 activation of the AKT pathway in primary myelofibrosis (PMF). Only JAK1/2 inhibitor ruxolitinib largely activated ERK1/2 signaling in essential thrombocythemia and PMF (up to 4.6 fold), with a more prominent activation in JAK2V617F positive granulocytes. Regarding a cell cycle, we found that IL-6 reduction of HEL cells percentage in G2M phase was reversed by ruxolitinib (2.6 fold). Moreover, ruxolitinib potentiated apoptosis of PMF granulocytes (1.6 fold). Regarding DNA replication, we found that ruxolitinib prevented the IL-6 augmentation of MPN granulocytes frequency in the S phase of the cell cycle (up to 2.9 fold). The inflammatory stimulation induces a cross-talk between the proliferation linked pathways, where JAK1/2 inhibition is compensated by the activation of the ERK1/2 pathway during IL-6 stimulation of DNA replication.


Assuntos
Replicação do DNA/efeitos dos fármacos , Interleucina-6/farmacologia , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transtornos Mieloproliferativos/patologia , Adulto , Idoso , Antígenos CD34/metabolismo , Linhagem Celular Tumoral , Feminino , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Humanos , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Masculino , Pessoa de Meia-Idade , Transtornos Mieloproliferativos/metabolismo , Nitrilas , Fosforilação/efeitos dos fármacos , Polimorfismo de Nucleotídeo Único , Pirazóis/farmacologia , Pirimidinas , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Fatores de Transcrição STAT/metabolismo
11.
Dev Dyn ; 247(3): 359-367, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28850772

RESUMO

Due to coexistence of stromal and epithelial tumor cells, their dynamic interactions have been widely recognized as significant cellular components to the tumor tissue integrity. Initiation and outcome of epithelial to mesenchymal transition (EMT) in tumor cells are dependent on their interaction with adjacent or recruited mesenchymal stromal cells (MSCs). A plethora of mechanisms are involved in MSCs-controlled employment of the developmental processes of EMT that contribute to loss of epithelial cell phenotype and acquisition of stemness, invasiveness and chemoresistance of tumor cells. Interplay of MSCs with tumor cells, including interchange of soluble biomolecules, plasma membrane structures, cytoplasmic content, and organelles, is established through cell-cell contact and/or by means of paracrine signaling. The main focus of this review is to summarize knowledge about involvement of MSCs in cancer cell EMT. Understanding the underlying cellular and molecular mechanism involved in the interplay between MSCs and cancer EMT is essential for development of effective therapy approaches, which in combination with current treatments may improve the control of tumor progression. Developmental Dynamics 247:359-367, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Transição Epitelial-Mesenquimal , Células-Tronco Mesenquimais/patologia , Neoplasias/patologia , Animais
12.
Dev Dyn ; 247(3): 382-395, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28722327

RESUMO

Transforming growth factor-ß (TGF-ß) is a pleiotropic factor that acts as a tumor suppressor in the early stages, while it exerts tumor promoting activities in advanced stages of cancer development. One of the hallmarks of cancer progression is the capacity of cancer cells to migrate and invade surrounding tissues with subsequent metastasis to different organs. Matrix metalloproteinases (MMPs) together with urokinase-type plasminogen activator (uPA) and its receptor (uPAR), whose main original function described is the proteolytic degradation of the extracellular matrix, play key cellular roles in the enhancement of cell malignancy during cancer progression. TGF-ß tightly regulates the expression of several MMPs and uPA/uPAR in cancer cells, which in return can participate in TGF-ß activation, thus contributing to tumor malignancy. TGF-ß is one of the master factors in the induction of cancer-associated epithelial to mesenchymal transition (EMT), and recently both MMPs and uPA/uPAR have also been shown to be implicated in the cancer-associated EMT process. In this review, we analyze the main molecular mechanisms underlying MMPs and uPA/uPAR regulation by TGF-ß, as well as their mutual implication in the development of EMT in cancer cells. Developmental Dynamics 247:382-395, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Transição Epitelial-Mesenquimal , Metaloproteinases da Matriz/metabolismo , Neoplasias/patologia , Fator de Crescimento Transformador beta/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Movimento Celular , Progressão da Doença , Matriz Extracelular/metabolismo , Humanos , Fator de Crescimento Transformador beta/metabolismo
13.
Adv Exp Med Biol ; 1107: 91-112, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30105601

RESUMO

In orthopedic medicine, a feasible reconstruction of bone structures remains one of the main challenges both for healthcare and for improvement of patients' quality of life. There is a growing interest in mesenchymal stem cells (MSCs) medical application, due to their multilineage differentiation potential, and tissue engineering integration to improve bone repair and regeneration. In this review we will describe the main characteristics of MSCs, such as osteogenesis, immunomodulation and antibacterial properties, key parameters to consider during bone repair strategies. Moreover, we describe the properties of calcium phosphate (CaP) bioceramics, which demonstrate to be useful tools in combination with MSCs, due to their biocompatibility, osseointegration and osteoconduction for bone repair and regeneration. Also, we overview the main characteristics of dental cavity MSCs, which are promising candidates, in combination with CaP bioceramics, for bone regeneration and tissue engineering. The understanding of MSCs biology and their interaction with CaP bioceramics and other biomaterials is critical for orthopedic surgical bone replacement, reconstruction and regeneration, which is an integrative and dynamic medical, scientific and bioengineering field of research and biotechnology.


Assuntos
Regeneração Óssea , Fosfatos de Cálcio/química , Cerâmica/química , Células-Tronco Mesenquimais/citologia , Diferenciação Celular , Humanos , Osteogênese , Engenharia Tecidual
14.
Biochim Biophys Acta ; 1853(2): 431-44, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25433194

RESUMO

Mesenchymal stem cells (MSCs) have the potential to migrate toward damaged tissues increasing tissue regeneration. Interleukin-17 (IL-17) is a proinflammatory cytokine with pleiotropic effects associated with many inflammatory diseases. Although IL-17 can modulate MSC functions, its capacity to regulate MSC migration is not well elucidated so far. Here, we studied the role of IL-17 on peripheral blood (PB) derived MSC migration and transmigration across endothelial cells. IL-17 increased PB-MSC migration in a wound healing assay as well as cell mobilization from collagen gel. Concomitantly IL-17 induced the expression of urokinase type plasminogen activator (uPA) without affecting matrix metalloproteinase expression. The incremented uPA expression mediated the capacity of IL-17 to enhance PB-MSC migration in a ERK1,2 MAPK dependent way. Also, IL-17 induced PB-MSC migration alongside with changes in cell polarization and uPA localization in cell protrusions. Moreover, IL-17 increased PB-MSC adhesion to endothelial cells and transendothelial migration, as well as increased the capacity of PB-MSC adhesion to fibronectin, in an uPA-dependent fashion. Therefore, our data suggested that IL-17 may act as chemotropic factor for PB-MSCs by incrementing cell motility and uPA expression during inflammation development.


Assuntos
Células Sanguíneas/citologia , Movimento Celular/efeitos dos fármacos , Interleucina-17/farmacologia , Células-Tronco Mesenquimais/citologia , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Células Sanguíneas/efeitos dos fármacos , Células Sanguíneas/enzimologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Polaridade Celular/efeitos dos fármacos , Colágeno/metabolismo , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibronectinas/metabolismo , Humanos , Imunofenotipagem , Metaloproteinases da Matriz/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/enzimologia , Camundongos , Receptores de Interleucina-17/metabolismo
15.
Cell Biol Int ; 39(5): 619-28, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25597879

RESUMO

Transforming growth factor ß (TGF-ß) modulates capacity of macrophages to produce urokinase type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP9). uPA and MMP9 actively participate in extracellular matrix reorganization and influence macrophages chemotaxis and cell migration. Although, TGF-ß regulates uPA and MMP9 macrophages expression, the underlying intracellular signal mechanisms are not well elucidated so far. Here we have investigated the implication of TGF-ß signaling in the regulation of uPA and MMP9 expression in RAW 264.7 macrophages. The expression of uPA and MMP9 was assessed by zymography, Western blotting and RT-PCR. The involvement of Smad, MAPK or NFκB signaling pathways was evaluated by using specific inhibitors. Our results indicated that TGF-ß simultaneously increased uPA and reduced MMP9 expression. The Smad3, ERK1,2, and JNK1,2 signaling pathways seem to be the main mechanisms that mediate TGF-ß-induced uPA expression. Whereas TGF-ß-reduced MMP9 expression appears to be regulated independently by JNK1,2 activation and by NFκB signaling inhibition. Thus, our results suggested that, in murine macrophages, TGF-ß differentially regulates uPA and MMP9 expressions through different intracellular signaling mechanisms. In addition, presented data may help in understanding the role of TGF-ß in macrophages proteases regulation in inflammatory diseases.


Assuntos
Macrófagos/efeitos dos fármacos , Metaloproteinase 9 da Matriz/genética , Fator de Crescimento Transformador beta/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
Mediators Inflamm ; 2015: 470458, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25999667

RESUMO

Adult stem cells have a great potential applicability in regenerative medicine and cell-based therapies. However, there are still many unresolved issues concerning their biology, and the influence of the local microenvironment on properties of stem cells has been increasingly recognized. Interleukin (IL-) 17, as a cytokine implicated in many physiological and pathological processes, should be taken into consideration as a part of a regulatory network governing tissue-associated stem cells' fate. This review is focusing on the published data on the effects of IL-17 on the properties and function of hematopoietic and mesenchymal stem cells and trying to discuss that IL-17 achieves many of its roles by acting on adult stem cells.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Interleucina-17/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos
17.
ScientificWorldJournal ; 2014: 521754, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24578639

RESUMO

Transforming growth factor-beta (TGF-ß) is a pleiotropic factor with several different roles in health and disease. In tumorigenesis, it may act as a protumorigenic factor and have a profound impact on the regulation of the immune system response. Matrix metalloproteinases (MMPs) are a family that comprises more than 25 members, which have recently been proposed as important regulators acting in tumor stroma by regulating the response of noncellular and cellular microenvironment. Tumor stroma consists of several types of resident cells and infiltrating cells derived from bone marrow, which together play crucial roles in the promotion of tumor growth and metastasis. In cancer cells, TGF-ß regulates MMPs expression, while MMPs, produced by either cancer cells or residents' stroma cells, activate latent TGF-ß in the extracellular matrix, together facilitating the enhancement of tumor progression. In this review we will focus on the compartment of myeloid stroma cells, such as tumor-associated macrophages, neutrophils, and dendritic and mast cells, which are potently regulated by TGF-ß and produce large amounts of MMPs. Their interplay and mutual implications in the generation of pro-tumorigenic cancer microenvironment will be analyzed.


Assuntos
Comunicação Celular , Metaloproteases/biossíntese , Células Mieloides/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral , Animais , Regulação Enzimológica da Expressão Gênica , Humanos , Células Mieloides/patologia , Neoplasias/patologia , Células Estromais/metabolismo , Células Estromais/patologia
18.
Acta Histochem ; 126(5-7): 152183, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39029317

RESUMO

Myeloid malignancies stem from a modified hematopoietic stem cell and predominantly include acute myeloid leukemia, myelodysplastic neoplasms, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid-derived suppressor cells (MDSCs) exhibit immunoregulatory properties by governing the innate and adaptive immune systems, creating a permissive and supportive environment for neoplasm growth. This review examines the key characteristics of MDSCs in myeloid malignancies, highlighting that an increased MDSC count corresponds to heightened immunosuppressive capabilities, fostering an immune-tolerant neoplasm microenvironment. Also, this review analyzes and describes the potential of combined cancer therapies, focusing on targeting MDSC generation, expansion, and their inherent immunosuppressive activities to enhance the efficacy of current cancer immunotherapies. A comprehensive understanding of the implications of myeloid malignancies may enhance the exploration of immunotherapeutic strategies for their potential application.

19.
Heliyon ; 10(3): e25081, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38314300

RESUMO

Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.

20.
Spectrochim Acta A Mol Biomol Spectrosc ; 322: 124824, 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39029203

RESUMO

In this study, we have reported a novel 4-bromo-salicylaldehyde-diphenyl-azine (B-1), a new member of salicylaldehyde-diphenyl-azine (SDPA) family known for its excellent sensing properties. In contrast to the previously reported AIEgens, we found that the bromo-substitution at the 4th position of the salicylaldehyde moiety blue-shifted the emission by 10 and 15 nm as compared to the unsubstituted (Tong et.al 2017) and Bromo at the 5th position (Jain et.al 2023) respectively. Moreover, B-1 crystallizes instantly as the cooling process starts, which was not observed in the previously reported scaffolds. The sensing investigation again demonstrated the precise and ultrasensitive behavior of B-1 for copper ions. B-1 has a very low LOD value i.e. 29.2 x 10-8 M with a high association constant and binds with copper ion in 2:1 mode. This time we also analyzed the practical applicability in the solid phase using cotton swabs and performed the real-time estimation of copper ions in water and biological samples like urine and blood serum. The excellent percentage recovery and the RSD value suggest the precision of the experiments. Further, we also perform the sensing in living cancer HeLa cells. Altogether, we found that the SDPA skeleton is precise and ultrasensitive for copper ions and versatile which can be used variously to detect copper ions in the real world. This research will surely help in developing new specific skeleton-based AIEgens with desirable emission properties and precise applications in the future.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa