Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 574
Filtrar
1.
Bioorg Med Chem ; 100: 117628, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38330850

RESUMO

Although neuroinflammation is a significant pathogenic feature of many neurologic disorders, its precise function in-vivo is still not completely known. PET imaging enables the longitudinal examination, quantification, and tracking of different neuroinflammation biomarkers in living subjects. Particularly, PET imaging of Microglia, specialised dynamic immune cells crucial for maintaining brain homeostasis in central nervous system (CNS), is crucial for staging the neuroinflammation. Colony Stimulating Factor- 1 Receptor (CSF-1R) PET imaging is a novel method for the quantification of neuroinflammation. CSF-1R is mainly expressed on microglia, and neurodegenerative disorders greatly up-regulate its expression. The present review primarily focuses on the development, pros and cons of all the CSF-1R PET tracers reported for neuroinflammation imaging. Apart from neuroinflammation imaging, CSF-1R inhibitors are also reported for the therapy of neurodegenerative diseases such as Alzheimer's disease (AD). AD is a prevalent, advancing, and fatal neurodegenerative condition that have the characteristic feature of persistent neuroinflammation and primarily affects the elderly. The aetiology of AD is profoundly influenced by amyloid-beta (Aß) plaques, intracellular neurofibrillary tangles, and microglial dysfunction. Increasing evidence suggests that CSF-1R inhibitors (CSF-1Ri) can be helpful in preclinical models of neurodegenerative diseases. This review article also summarises the most recent developments of CSF-1Ri-based therapy for AD.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Idoso , Humanos , Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Microglia/metabolismo , Doenças Neurodegenerativas/diagnóstico por imagem , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Doenças Neuroinflamatórias , Tomografia por Emissão de Pósitrons/métodos , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
2.
J Biol Chem ; 299(6): 104755, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37116708

RESUMO

The colony-stimulating factor 3 receptor (CSF3R) controls the growth of neutrophils, the most abundant type of white blood cell. In healthy neutrophils, signaling is dependent on CSF3R binding to its ligand, CSF3. A single amino acid mutation in CSF3R, T618I, instead allows for constitutive, ligand-independent cell growth and leads to a rare type of cancer called chronic neutrophilic leukemia. However, the disease mechanism is not well understood. Here, we investigated why this threonine to isoleucine substitution is the predominant mutation in chronic neutrophilic leukemia and how it leads to uncontrolled neutrophil growth. Using protein domain mapping, we demonstrated that the single CSF3R domain containing residue 618 is sufficient for ligand-independent activity. We then applied an unbiased mutational screening strategy focused on this domain and found that activating mutations are enriched at sites normally occupied by asparagine, threonine, and serine residues-the three amino acids which are commonly glycosylated. We confirmed glycosylation at multiple CSF3R residues by mass spectrometry, including the presence of GalNAc and Gal-GalNAc glycans at WT threonine 618. Using the same approach applied to other cell surface receptors, we identified an activating mutation, S489F, in the interleukin-31 receptor alpha chain. Combined, these results suggest a role for glycosylated hotspot residues in regulating receptor signaling, mutation of which can lead to ligand-independent, uncontrolled activity and human disease.


Assuntos
Leucemia Neutrofílica Crônica , Humanos , Leucemia Neutrofílica Crônica/diagnóstico , Leucemia Neutrofílica Crônica/genética , Leucemia Neutrofílica Crônica/metabolismo , Glicosilação , Ligantes , Mutação , Receptores de Fator Estimulador de Colônias/genética , Receptores de Fator Estimulador de Colônias/metabolismo , Treonina/metabolismo , Fatores Estimuladores de Colônias/genética , Fatores Estimuladores de Colônias/metabolismo
3.
BMC Cancer ; 23(1): 251, 2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36922758

RESUMO

BACKGROUND: Currently there are no established fertility preservation options for pre-pubertal boys facing cancer treatment. Granulocyte-colony stimulating factor (G-CSF) treatment has been proposed to be chemoprotective against spermatogonial cell loss in an alkylating chemotherapy model of busulfan treated adult mice. Having previously shown that exposure to the alkylating-like chemotherapy cisplatin resulted in a reduction in germ cell numbers in immature human testicular tissues, we here investigate whether G-CSF would prevent cisplatin-induced germ cell loss in immature human and mouse (fetal and pre-pubertal) testicular tissues. METHODS: Organotypic in vitro culture systems were utilised to determine the effects of clinically-relevant concentrations of G-CSF in cisplatin-exposed immature testicular tissues. Human fetal (n = 14 fetuses) and mouse pre-pubertal (n = 4 litters) testicular tissue pieces were cultured and exposed to cisplatin or vehicle control for 24 hrs and analysed at 72 and 240 hrs post-exposure. Combined G-CSF and cisplatin exposure groups explored varying concentrations and duration of G-CSF supplementation to the culture medium (including groups receiving G-CSF before, during and after cisplatin exposure). In addition, effects of G-CSF supplementation alone were investigated. Survival of total germ cell and sub-populations were identified by expression of AP2γ and MAGE-A4 for human gonocytes and (pre)spermatogonia, respectively, and MVH and PLZF, for mouse germ cells and putative spermatogonial stem cells (SSCs) respectively, were quantified. RESULTS: Exposure to cisplatin resulted in a reduced germ cell number in human fetal and mouse pre-pubertal testicular tissues at 240 hrs post-exposure. Germ cell number was not preserved by combined exposure with G-CSF using any of the exposure regimens (prior to, during or after cisplatin exposure). Continuous supplementation with G-CSF alone for 14 days did not change the germ cell composition in either human or mouse immature testicular tissues. CONCLUSIONS: This study demonstrates that exposure to G-CSF does not prevent cisplatin-induced germ cell loss in immature human and mouse testicular tissues in an in vitro system.


Assuntos
Cisplatino , Testículo , Masculino , Humanos , Animais , Camundongos , Testículo/metabolismo , Cisplatino/farmacologia , Espermatogônias , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Fatores Estimuladores de Colônias/farmacologia , Granulócitos
4.
Sci Rep ; 12(1): 21663, 2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36522405

RESUMO

The human type II collagen (Col II), specifically expressed in chondrocytes, is a crucial component of the adult hyaline cartilage. We examine the potential of artificial induction of Col II in human peripheral blood mononuclear cells (PBMNCs) as a novel Col II provider. Human PBMNCs were purified and were treated with high doses of macrophage-colony stimulating factor (M-CSF), granulocyte macrophage-colony stimulating factor (GM-CSF), or granulocyte-colony stimulating factor (G-CSF) and examined the Col II expression at indicated days. Quantitative Col II expression was validated by real-time reverse transcriptase-polymerase chain reaction (RT-PCR), immunocytochemistry, and flow cytometry. We demonstrate that monocytes in PBMNCs can be artificially induced to express both Col II proteins and M2 macrophage markers by the high concentration of colony-stimulating factors, especially M-CSF and GM-CSF. The Col II proteins were detected on the cell membrane and in the cytoplasm by flow cytometry and immunocytostaining. Combination with IL-4 provided a synergistic effect with M-CSF/GM-CSF to trigger Col II expression in M2 macrophages. These CD206 and Col II double-expressing cells, named modified macrophages, share M2 macrophages' anti-inflammatory potency. We demonstrated that the modified macrophages could significantly attenuate the inflammatory progress of Complete Freund's adjuvant (CFA)-induced arthritis and collagen-induced arthritis in rodents. Here, we provide the first evidence that a modified macrophage population could ectopically express Col II and control the progress of arthritis in animals.


Assuntos
Artrite Experimental , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Animais , Humanos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Leucócitos Mononucleares/metabolismo , Macrófagos/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Artrite Experimental/metabolismo
5.
J Neurochem ; 160(6): 643-661, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34935149

RESUMO

Multiple sclerosis (MS), especially in its progressive phase, involves early axonal and neuronal damage resulting from a combination of inflammatory mediators, demyelination, and loss of trophic support. During progressive disease stages, a microenvironment is created within the central nervous system (CNS) favoring the arrival and retention of inflammatory cells. Active demyelination and neurodegeneration have also been linked to microglia (MG) and astrocyte (AST)-activation in early lesions. While reactive MG can damage tissue, exacerbate deleterious effects, and contribute to neurodegeneration, it should be noted that activated MG possess neuroprotective functions as well, including debris phagocytosis and growth factor secretion. The progressive form of MS can be modeled by the prolonged administration to cuprizone (CPZ) in adult mice, as CPZ induces highly reproducible demyelination of different brain regions through oligodendrocyte (OLG) apoptosis, accompanied by MG and AST activation and axonal damage. Therefore, our goal was to evaluate the effects of a reduction in microglial activation through orally administered brain-penetrant colony-stimulating factor-1 receptor (CSF-1R) inhibitor BLZ945 (BLZ) on neurodegeneration and its correlation with demyelination, astroglial activation, and behavior in a chronic CPZ-induced demyelination model. Our results show that BLZ treatment successfully reduced the microglial population and myelin loss. However, no correlation was found between myelin preservation and neurodegeneration, as axonal degeneration was more prominent upon BLZ treatment. Concomitantly, BLZ failed to significantly offset CPZ-induced astroglial activation and behavioral alterations. These results should be taken into account when proposing the modulation of microglial activation in the design of therapies relevant for demyelinating diseases. Cover Image for this issue: https://doi.org/10.1111/jnc.15394.


Assuntos
Doenças Desmielinizantes , Esclerose Múltipla , Animais , Fatores Estimuladores de Colônias/efeitos adversos , Fatores Estimuladores de Colônias/metabolismo , Cuprizona/metabolismo , Cuprizona/toxicidade , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo
6.
Am J Respir Cell Mol Biol ; 64(3): 318-330, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33264084

RESUMO

Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-ß-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvß3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Pulmão/irrigação sanguínea , Pulmão/crescimento & desenvolvimento , NF-kappa B/metabolismo , Neovascularização Fisiológica , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Movimento Celular , Fatores Estimuladores de Colônias/metabolismo , Células Endoteliais/metabolismo , Integrina alfaVbeta3/metabolismo , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Nascimento Prematuro , Alvéolos Pulmonares/metabolismo , Ovinos
7.
Probl Radiac Med Radiobiol ; 25: 456-477, 2020 Dec.
Artigo em Inglês, Ucraniano | MEDLINE | ID: mdl-33361854

RESUMO

OBJECTIVE: to establish the connection of radiation-induced changes in gene expression with the realized pathology of the broncho-pulmonary and cardiovascular systems in Chornobyl clean-up workers. MATERIALS AND METHODS: We examined 314 male Chornobyl clean-up workers (main group; age (58.94 ± 6.82) years(M ± SD); min 33, max 79 years; radiation dose (411.82 ± 625.41) mSv (M ± SD); min 1.74, max 3600 mSv) with various nosological forms of cardiovascular and broncho-pulmonary pathology (BPP) and 50 subjects of the controlgroup: age (50.50 ± 5.73) years (M ± SD); min 41, max 67 years. The relative level of BCL2, CDKN2A, CLSTN2, GSTM1,IFNG, IL1B, MCF2L, SERPINB9, STAT3, TERF1, TERF2, TERT, TNF, TP53, CCND1, CSF2, VEGFA genes expression was determined inperipheral blood leukocytes by real-time PCR (7900 HT Fast Real-Time PCR System (Applied Biosystems, USA)). The«gene-disease¼ association was determined on statistical models stratified separately for each disease and gene.Logistic regression was used to calculate the odds ratio. RESULTS: Increased GSTM1 gene expression and no changes in angiogenesis-related VEGFA gene expression werefound in the main group of patients with coronary heart disease (CHD). It was established overexpression of TP53,VEGF and IFNG genes in the group of patients with arterial hypertension (AH). At combination of these diseases anincrease of expression of СSF2, TERF1, TERF2 genes was established. The detected changes demonstrate an activationof the antioxidative defense system in patients with CHD, while AH is associated with the expression of genes ofangiogenesis and immune inflammation. It was shown an increase in the expression of genes associated with apoptosis and kinase activity (BCL2, CLSTN2, CDKN2), immune inflammation (CSF2, IL1B, TNF) in Chornobyl clean-upworkers with BPP. Expression of TP53 and GSTM1 (gene, associated with the glutathione system) was significantlyupregulated in the group of individuals with chronic bronchitis, whereas in patients with chronic obstructive pulmonary disease, no increase was detected; the expression of SERPINB9 and MCF2L genes was downregulated. CONCLUSIONS: Changes in the expression of genes, associated with the development of somatic pathology in theremote period after irradiation, in particular the genes of the immune response and inflammatory reactions CSF2,IFNG, IL1B, TNF; expression of genes that regulate cell proliferation, aging and apoptosis TP53, BCL2, MCF2L, CDKN2A,SERPINB9, TERF1, TERF2, TERT; genes that regulate cell adhesion and angiogenesis CLSTN2, VEGF.


Assuntos
Doenças Cardiovasculares/genética , Acidente Nuclear de Chernobyl , Expressão Gênica/efeitos da radiação , Pneumopatias/genética , Exposição à Radiação/efeitos adversos , Lesões por Radiação/genética , Idoso , Poluentes Radioativos do Ar/efeitos adversos , Proteínas Angiogênicas/genética , Proteínas Angiogênicas/metabolismo , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Fatores Estimuladores de Colônias/genética , Fatores Estimuladores de Colônias/metabolismo , Socorristas , Contaminação Radioativa de Alimentos , Humanos , Interferons/genética , Interferons/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Leucócitos Mononucleares/efeitos da radiação , Pneumopatias/etiologia , Pneumopatias/metabolismo , Pneumopatias/patologia , Masculino , Pessoa de Meia-Idade , Prognóstico , Lesões por Radiação/etiologia , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Radioisótopos , Poluentes Radioativos do Solo/efeitos adversos , Proteínas de Ligação a Telômeros/genética , Proteínas de Ligação a Telômeros/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ucrânia
8.
Dev Comp Immunol ; 113: 103798, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32745480

RESUMO

The differentiation of distinct leukocyte subsets is governed by lineage-specific growth factors that elicit disparate expression of transcription factors and markers by the developing cell populations. For example, macrophages (Mφs) and granulocytes (Grns) arise from common granulocyte-macrophage progenitors in response to distinct myeloid growth factors. In turn, myelopoiesis of the Xenopus laevis anuran amphibian appears to be unique to other studied vertebrates in several respects while the functional differentiation of amphibian Mφs and Grns from their progenitor cells remains poorly understood. Notably, the expression of colony stimulating factor-1 receptor (CSF-1R) or CSF-3R on granulocyte-macrophage progenitors marks their commitment to Mφ- or Grn-lineages, respectively. CSF-1R is activated by the colony stimulating factor-1 (CSF-1) and interleukin (IL-34) cytokines, resulting in morphologically and functionally distinct Mφ cell types. Conversely, CSF-3R is ligated by CSF-3 in a process indispensable for granulopoiesis. Presently, we explore the relationships between X. laevis CSF-1-Mφs, IL-34-Mφs and CSF-3-Grns by examining their expression of key lineage-specific transcription factor and myeloid marker genes as well as their enzymology. Our findings suggest that while the CSF-1- and IL-34-Mφs share some commonalities, the IL-34-Mφs possess transcriptional patterns more akin to the CSF-3-Grns. IL-34-Mφs also possess robust expression of dendritic cell-associated transcription factors and surface marker genes, further underlining the difference between this cell type and the CSF-1-derived frog Mφ subset. Moreover, the three myeloid populations differ in their respective tartrate-resistant acid phosphatase, specific- and non-specific esterase activity. Together, this work grants new insights into the developmental relatedness of these three frog myeloid subsets.


Assuntos
Granulócitos/fisiologia , Macrófagos/fisiologia , Xenopus laevis/imunologia , Proteínas de Anfíbios/genética , Proteínas de Anfíbios/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Linhagem da Célula , Fatores Estimuladores de Colônias/metabolismo , Esterases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Interleucinas/genética , Interleucinas/metabolismo , Mielopoese , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Transcriptoma
9.
Gene ; 760: 145003, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32739587

RESUMO

Imiquimod (IMQ) is approved as a first-line treatment for genital warts caused by human papillomavirus (HPV) infection. However, the recurrence rate is very high. HPV E7 protein plays a critical role in HPV immune escape. However, the role of HPV11 E7 protein in genital warts recurrence during IMQ treatment is not clear. Here, we found that the expression profile of NHEK cells was obviously changed after IMQ treatment, and a large number of genes encoding cytokines and genes involved in cytokine-mediated signaling pathways and cellular metabolic signaling pathways were up- or downregulated. HPV11E7 overexpression inhibited the IMQ-induced production of of multiple chemokines and colony-stimulating factors in NHEK cells. Furthermore, we found that HPV11E7 could impair the activation of mitogen-activated protein kinase (MAPK) signaling pathway. Therefore, our results suggested that HPV11 E7 diminishes the production of chemokines, colony-stimulating factors and other cytokines via inhibition of the MAPK signaling pathway, which suppresses the therapeutic effect of IMQ and promotes the recurrence of diseases, such as condyloma acuminatum.


Assuntos
Imiquimode/farmacologia , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Quimiocinas/biossíntese , Quimiocinas/genética , Quimiocinas/metabolismo , Fatores Estimuladores de Colônias/biossíntese , Fatores Estimuladores de Colônias/metabolismo , Citocinas/metabolismo , Expressão Gênica/efeitos dos fármacos , Papillomavirus Humano 11/metabolismo , Humanos , Imiquimode/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Oncogênicas Virais/imunologia , Infecções por Papillomavirus/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
10.
Dev Comp Immunol ; 108: 103671, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32147469

RESUMO

Interleukin-3 (IL-3), IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF) are related cytokines that signal through receptors possessing the ß common (ßc) chain. As a family, these cytokines combine rather non-specific hematopoietic growth factor properties with a special importance for eosinophils, basophils, and mast cells. In fish the cytokines of this family are called IL-5fam, and the present study, using carp, constitutes their first functional analysis. Carp il-5fam expression was enhanced by stimulation with phytohemagglutinin and killed bacteria. Reminiscent of mammalian IL-3/IL-5/GM-CSF family members, recombinant carp IL-5fam (rcIL-5fam) induced activation of transcription factor STAT5 and efficiently promoted proliferation and colony-formation of eosinophil/basophil/mast-cell type (EBM) granulocytes. Upon addition of recombinant carp ßc the growth effect of rcIL-5fam was reduced, suggesting ßc participation in the signaling route. In summary, despite differences in individual cytokines and cell populations, fish and mammalian IL-3/IL-5/GM-CSF family members share growth factor functions for non-neutrophil granulocytes.


Assuntos
Carpas/imunologia , Fatores Estimuladores de Colônias/metabolismo , Proteínas de Peixes/metabolismo , Granulócitos/imunologia , Interleucinas/metabolismo , Animais , Carpas/genética , Carpas/metabolismo , Carpas/microbiologia , Proliferação de Células , Fatores Estimuladores de Colônias/genética , Fatores Estimuladores de Colônias/isolamento & purificação , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/genética , Proteínas de Peixes/isolamento & purificação , Granulócitos/metabolismo , Interleucinas/genética , Interleucinas/isolamento & purificação , Fito-Hemaglutininas/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT5/metabolismo
11.
Int J Biol Macromol ; 156: 204-216, 2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32156537

RESUMO

Calf thymus polypeptide (CTP) is prepared from calf thymus. It has a molecular mass of <10 kilodalton (kDa) and contains 17 types of amino acids. This study investigated the hematopoietic function-improvement effect of CTP in CHRF, K562, and bone marrow mononuclear cells; mice with immunosuppression; and with hematopoietic dysfunction. In mice with immunosuppression, CTP enhanced the cytotoxic activity of natural killer cells and the proliferation of lymphocytes and regulated the levels of immunoglobulins. It also enhanced the proliferation and differentiation of CHRF and K562 cells by upregulating the expression of proliferation- and differentiation-related proteins. In mice with hematopoietic dysfunction, CTP restored white blood cell, neutrophil, and hemoglobin proportions in the peripheral blood and enhanced the levels of B lymphocytes and hematopoietic stem cells and progenitor cells in the bone marrow. CTP effectively regulated the levels of hematopoiesis-related cytokines, such as granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), interleukin 2, and interferons-γ, and enhanced the expression of hematopoiesis-related proteins in both primary bone marrow cells and mice with hematopoietic dysfunction. These results indicate that CTP has hematopoietic function-improvement effect and this effect may be related to the modulation of colony-stimulating factors (CSFs) and related signaling pathways.


Assuntos
Fatores Estimuladores de Colônias/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Peptídeos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Biomarcadores , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Bovinos , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Fatores Estimuladores de Colônias/metabolismo , Citocinas/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hospedeiro Imunocomprometido , Fatores Imunológicos/química , Fatores Imunológicos/farmacologia , Imunomodulação/efeitos dos fármacos , Imunofenotipagem , Ativação Linfocitária , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peso Molecular , Especificidade de Órgãos/imunologia , Peptídeos/química , Timo/metabolismo
12.
Nat Microbiol ; 4(12): 2273-2284, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31548683

RESUMO

The deoxynucleotide triphosphate (dNTP) hydrolase SAMHD1 inhibits retroviruses in non-dividing myeloid cells. Although antiviral activity towards DNA viruses has also been demonstrated, the role of SAMHD1 during cytomegalovirus (CMV) infection remains unclear. To determine the impact of SAMHD1 on the replication of CMV, we used murine CMV (MCMV) to infect a previously established SAMHD1 knockout mouse model and found that SAMHD1 inhibits the replication of MCMV in vivo. By comparing the replication of MCMV in vitro in myeloid cells and fibroblasts from SAMHD1-knockout and control mice, we found that the viral kinase M97 counteracts SAMHD1 after infection by phosphorylating the regulatory residue threonine 603. The phosphorylation of SAMHD1 in infected cells correlated with a reduced level of dNTP hydrolase activity and the loss of viral restriction. Together, we demonstrate that SAMHD1 acts as a restriction factor in vivo and we identify the M97-mediated phosphorylation of SAMHD1 as a previously undescribed viral countermeasure.


Assuntos
Muromegalovirus/efeitos dos fármacos , Fosfotransferases/metabolismo , Proteína 1 com Domínio SAM e Domínio HD/antagonistas & inibidores , Proteína 1 com Domínio SAM e Domínio HD/metabolismo , Animais , Antivirais/farmacologia , Fatores Estimuladores de Colônias/metabolismo , Modelos Animais de Doenças , Células HEK293 , Infecções por Herpesviridae/tratamento farmacológico , Infecções por Herpesviridae/virologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muromegalovirus/enzimologia , Muromegalovirus/crescimento & desenvolvimento , Células NIH 3T3 , Fosforilação , Proteínas Recombinantes , Proteína 1 com Domínio SAM e Domínio HD/genética , Transcriptoma , Proteínas Virais/metabolismo , Replicação Viral/efeitos dos fármacos
13.
PLoS Biol ; 17(3): e2006859, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30921319

RESUMO

Brain metastases are prevalent in various types of cancer and are often terminal, given the low efficacy of available therapies. Therefore, preventing them is of utmost clinical relevance, and prophylactic treatments are perhaps the most efficient strategy. Here, we show that systemic prophylactic administration of a toll-like receptor (TLR) 9 agonist, CpG-C, is effective against brain metastases. Acute and chronic systemic administration of CpG-C reduced tumor cell seeding and growth in the brain in three tumor models in mice, including metastasis of human and mouse lung cancer, and spontaneous melanoma-derived brain metastasis. Studying mechanisms underlying the therapeutic effects of CpG-C, we found that in the brain, unlike in the periphery, natural killer (NK) cells and monocytes are not involved in controlling metastasis. Next, we demonstrated that the systemically administered CpG-C is taken up by endothelial cells, astrocytes, and microglia, without affecting blood-brain barrier (BBB) integrity and tumor brain extravasation. In vitro assays pointed to microglia, but not astrocytes, as mediators of CpG- C effects through increased tumor killing and phagocytosis, mediated by direct microglia-tumor contact. In vivo, CpG-C-activated microglia displayed elevated mRNA expression levels of apoptosis-inducing and phagocytosis-related genes. Intravital imaging showed that CpG-C-activated microglia cells contact, kill, and phagocytize tumor cells in the early stages of tumor brain invasion more than nonactivated microglia. Blocking in vivo activation of microglia with minocycline, and depletion of microglia with a colony-stimulating factor 1 inhibitor, indicated that microglia mediate the antitumor effects of CpG-C. Overall, the results suggest prophylactic CpG-C treatment as a new intervention against brain metastasis, through an essential activation of microglia.


Assuntos
Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/metabolismo , Microglia/metabolismo , Microglia/patologia , Oligodesoxirribonucleotídeos/uso terapêutico , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Fatores Estimuladores de Colônias/antagonistas & inibidores , Fatores Estimuladores de Colônias/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/metabolismo , Masculino , Melanoma/complicações , Melanoma/metabolismo , Camundongos , Minociclina/metabolismo , Fagocitose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
14.
Eur Neuropsychopharmacol ; 29(3): 384-396, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30630651

RESUMO

Cognitive deterioration and symptom progression occur in schizophrenia over the course of the disorder. A dysfunction of the immune system/neuroinflammatory pathways has been linked to schizophrenia (SZ). These altered processes in the dorsolateral prefrontal cortex (DLPFC) could contribute to the worsening of the deficits. However, limited studies are available in this brain region in elderly population with long-term treatments. In this study, we explore the possible deregulation of 21 key genes involved in immune homeostasis, including pro- and anti-inflammatory cytokines, cytokine modulators (toll-like receptors, colony-stimulating factors, and members of the complement system) and microglial and astroglial markers in the DLPFC in elderly chronic schizophrenia. We used quantitative real-time reverse transcriptase polymerase chain reaction (RT-PCR) on extracts from postmortem DLPFC of elderly subjects with chronic SZ (n = 14) compared to healthy control individuals (n = 14). We report that CSF1R, TLR4, IL6, TNFα, TNFRSF1A, IL10, IL10RA, IL10RB, and CD68 were down-regulated in elderly SZ subjects. Moreover, we found that the expression levels of all the altered inflammatory genes in SZ correlated with the microglial marker CD68. However, no associations were found with the astroglial marker GFAP. This study reveals a decrease in the gene expression of cytokines and immune response/inflammation mediators in the DLPFC of elderly subjects with chronic schizophrenia, supporting the idea of a dysfunction of these processes in aged patients and its possible relationship with active microglia abundance. These findings include elements that might contribute to the cognitive decline and symptom progression linked to DLPFC functioning at advanced stages of the disease.


Assuntos
Citocinas/metabolismo , Regulação para Baixo/fisiologia , Encefalite/complicações , Córtex Pré-Frontal/metabolismo , Esquizofrenia/complicações , Esquizofrenia/patologia , Idoso , Idoso de 80 Anos ou mais , Fatores Estimuladores de Colônias/genética , Fatores Estimuladores de Colônias/metabolismo , Citocinas/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
15.
Proc Natl Acad Sci U S A ; 116(4): 1361-1369, 2019 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-30545915

RESUMO

Interleukin-1ß (IL-1ß) is abundant in the tumor microenvironment, where this cytokine can promote tumor growth, but also antitumor activities. We studied IL-1ß during early tumor progression using a model of orthotopically introduced 4T1 breast cancer cells. Whereas there is tumor progression and spontaneous metastasis in wild-type (WT) mice, in IL-1ß-deficient mice, tumors begin to grow but subsequently regress. This change is due to recruitment and differentiation of inflammatory monocytes in the tumor microenvironment. In WT mice, macrophages heavily infiltrate tumors, but in IL-1ß-deficient mice, low levels of the chemokine CCL2 hamper recruitment of monocytes and, together with low levels of colony-stimulating factor-1 (CSF-1), inhibit their differentiation into macrophages. The low levels of macrophages in IL-1ß-deficient mice result in a relatively high percentage of CD11b+ dendritic cells (DCs) in the tumors. In WT mice, IL-10 secretion from macrophages is dominant and induces immunosuppression and tumor progression; in contrast, in IL-1ß-deficient mice, IL-12 secretion by CD11b+ DCs prevails and supports antitumor immunity. The antitumor immunity in IL-1ß-deficient mice includes activated CD8+ lymphocytes expressing IFN-γ, TNF-α, and granzyme B; these cells infiltrate tumors and induce regression. WT mice with 4T1 tumors were treated with either anti-IL-1ß or anti-PD-1 Abs, each of which resulted in partial growth inhibition. However, treating mice first with anti-IL-1ß Abs followed by anti-PD-1 Abs completely abrogated tumor progression. These data define microenvironmental IL-1ß as a master cytokine in tumor progression. In addition to reducing tumor progression, blocking IL-1ß facilitates checkpoint inhibition.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Interleucina-1beta/antagonistas & inibidores , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antígeno CD11b/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Fatores Estimuladores de Colônias/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Feminino , Granzimas/farmacologia , Humanos , Terapia de Imunossupressão/métodos , Inflamação/metabolismo , Interferon gama/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
16.
Cytokine ; 122: 154190, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29074034

RESUMO

Colony stimulating factors (CSFs) play a central role in the development and functional maturation of immune cells besides having pleiotropic effects on cells of the vascular wall. The production of CSFs is induced by multiple atherogenic and inflammatory stimuli and their expression levels are often correlated positively with advanced atherosclerotic plaques and adverse cardiovascular events in humans suggesting that CSFs play a critical role in the pathophysiology of atherosclerosis progression. Interestingly, recombinant CSFs as well as anti-CSFs are being increasingly used for diverse clinical indications. However, the effect of these novel therapeutics on atherosclerotic plaque progression is not well understood. Herein, we summarize the currently available literature on the complex role of CSFs in various stages of atherosclerosis and emphasize the necessity for conducting further mechanistic studies in animal models of atherosclerosis as well as the need for evaluating the cardiovascular safety of CSF-based therapies in humans.


Assuntos
Aterosclerose/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator Estimulador de Colônias de Macrófagos/metabolismo , Animais , Aterosclerose/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fatores Estimuladores de Colônias/metabolismo , Fatores Estimuladores de Colônias/farmacologia , Progressão da Doença , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-3/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Modelos Animais , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/metabolismo
17.
Cytokine ; 111: 334-341, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30269030

RESUMO

BACKGROUND: Unregulated inflammatory and thrombotic responses have been proposed to be important causes of early brain injury and worse clinical outcomes after subarachnoid hemorrhage (SAH). OBJECTIVE: We hypothesize that SAH is characterized by an increased inflammatory and thrombotic state and disruption of associations between these states. METHODS: This is a retrospective cohort study of 60 patients with SAH. 23 patients with unruptured aneurysms (UA) and 77 patients with traumatic brain injury (TBI) were chosen as controls. Plasma cytokine levels were measured using a 41-plex human immunoassay kit, and cytokine patterns associated with SAH, UA and TBI were identified using statistical and informatics methods. RESULTS: SAH was characterized by an increase in several cytokines and chemokines, platelet-derived factors, and growth factors. Cluster analysis identified several cytokine clusters common in SAH, UA and TBI groups - generally grouped as platelet-derived, vascular and pro-inflammatory clusters. In the UA group, the platelet-derived cluster had an inverse relationship with the inflammatory cluster which was absent in SAH. Additionally, a cluster comprising of growth and colony stimulating factors was unique to SAH. CONCLUSIONS: A cluster of cytokines involved in growth and colony stimulation was unique to SAH. Negative associations between the thrombotic and inflammatory molecules were observed in UA but not in SAH. Further studies to examine the pathophysiology behind the cluster unique to SAH and the associations between the thrombotic and inflammatory cytokines are required.


Assuntos
Citocinas/metabolismo , Inflamação/metabolismo , Hemorragia Subaracnóidea/metabolismo , Plaquetas/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
18.
Dev Comp Immunol ; 84: 181-192, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29408047

RESUMO

Considering that macrophage functions are strongly impacted by the local tissue environment and the type of immune response, the aim of this study was to carefully set the methodological baseline for phenotype and functions of polarized porcine monocyte-derived macrophages. To this end, macrophages were generated in autologous serum alone or with colony-stimulating factor (CSF)-1 or CSF-2, and subsequently polarized with interferon (IFN)γ, interleukin-4 or IFNß. IFNγ promoted expression of MHC class I, MHC class II, CD11a, and CD40 as well as LPS-induced IL-6 and IL-12. A hallmark of interleukin-4 was Arginase 1 and CD203a upregulation, without abrogating pro-inflammatory cytokine production. IFNß induced CD169, MHC class I, CD40, CD80/86, but suppressed IL-6, IL-12 and tumor-necrosis-factor secretion. CSF-2 alone altered macrophage differentiation and promoted an IFNγ-like polarization. Altogether, the results provide a comprehensive overview of porcine macrophage polarization, and demonstrate commonalities with other species as well as peculiarities of the pig.


Assuntos
Interferon beta/metabolismo , Interferon gama/metabolismo , Macrófagos/imunologia , Monócitos/imunologia , Suínos/imunologia , Animais , Antígenos CD/metabolismo , Arginase/metabolismo , Diferenciação Celular , Células Cultivadas , Fatores Estimuladores de Colônias/metabolismo , Citocinas/metabolismo , Antígenos de Histocompatibilidade/metabolismo , Imunofenotipagem , Mediadores da Inflamação/metabolismo , Interleucina-4/metabolismo , Especificidade da Espécie
19.
Brain Behav Immun ; 68: 248-260, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29080683

RESUMO

Accumulation of microglia occurs in the dorsal horn in the rodent model of chronic post ischemic pain (CPIP), while the mechanism how microglia affects the development of persistent pain largely remains unknown. Here, using a rodent model of CPIP induced by ischemia-reperfusion (IR) injury in the hindpaw, we observed that microglial accumulation occurred in the ipsilateral dorsal horn after ischemia 3h, and in ipsilateral and contralateral dorsal horn in the rats with ischemia 6h. The accumulated microglia released BDNF, increased neuronal excitability in dorsal horn, and produced pain behaviors in the modeled rodents. We also found significantly increased signaling mediated by astrocytic colony-stimulating factor-1 (CSF1) and microglial CSF1 receptor (CSF1R) in dorsal horn in the ischemia 6h modeled rats. While exogenous M-CSF induced microglial activation and proliferation, BDNF production, neuronal hyperactivity in dorsal horn and behavioral hypersensitivity in the naïve rats, inhibition of astrocytic CSF1/microglial CSF1R signaling by fluorocitric or PLX3397 significantly suppressed microglial activation and proliferation, BDNF upregulation, and neuronal activity in dorsal horn, as well as the mechanical allodynia and thermal hyperalgesia, in the rats with ischemia 6h. Collectively, these results demonstrated that glial CSF1/CSF1R pathway mediated the microglial activation and proliferation, which facilitated the nociceptive output and contributed to the chronic pain induced by IR injury.


Assuntos
Dor Crônica/metabolismo , Fatores Estimuladores de Colônias/metabolismo , Receptores de Fator Estimulador de Colônias/metabolismo , Animais , Astrócitos/metabolismo , Comportamento Animal/fisiologia , Sensibilização do Sistema Nervoso Central/fisiologia , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hiperalgesia/metabolismo , Isquemia/fisiopatologia , Fator Estimulador de Colônias de Macrófagos/metabolismo , Masculino , Microglia/metabolismo , Neuralgia/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Regulação para Cima
20.
J Cell Physiol ; 233(2): 1500-1511, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28574591

RESUMO

In bone marrow (BM), hematopoietic elements are mingled with adipocytes (BM-A), which are the most abundant stromal component in the niche. BM-A progressively increase with aging, eventually occupying up to 50% of BM cavities. In this work, the role played by BM-A was explored by studying primary human BM-A isolated from hip surgery patients at the molecular level, through microarray analysis, and at the functional level, by assessing their relationship with primary human hematopoietic stem cells (HSC) by the long-term culture initiating cell (LTC-IC) assay. Findings demonstrated that BM-A are capable of supporting HSC survival in the LTC-IC assay, since after 5 weeks of co-culture, HSC were still able to proliferate and differentiate. Furthermore, critical molecules such as C-X-C motif chemokine 12 (CXCL12), interleukin (IL)-8, colony-stimulating factor 3 (CSF3), and leukaemia inhibitory factor (LIF), were expressed at similar levels in BM-A and in primary human BM mesenchymal stromal cells (BM-MSC), whereas IL-3 was higher in BM-A. Interestingly, BM-A displayed a different gene expression profile compared with subcutaneous adipose tissue adipocytes (AT-A) collected from abdominal surgery patients, especially in terms of regulation of lipid metabolism, stemness genes, and white-to-brown differentiation pathways. Accordingly, analysis of the gene pathways involved in hematopoiesis regulation showed that BM-A are more closely related to BM-MSC than to AT-A. The present data suggest that BM-A play a supporting role in the hematopoietic niche and directly sustain HSC survival.


Assuntos
Adipócitos/fisiologia , Células da Medula Óssea/fisiologia , Comunicação Celular , Células-Tronco Hematopoéticas/fisiologia , Adipócitos/metabolismo , Idoso , Idoso de 80 Anos ou mais , Células da Medula Óssea/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Técnicas de Cocultura , Fatores Estimuladores de Colônias/metabolismo , Feminino , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Humanos , Interleucina-8/metabolismo , Fator Inibidor de Leucemia/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Transdução de Sinais , Nicho de Células-Tronco , Gordura Subcutânea/citologia , Gordura Subcutânea/fisiologia , Fatores de Tempo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...