Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
Methods Mol Biol ; 2807: 271-283, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38743235

RESUMO

The blood-brain barrier (BBB) is one of several barriers between the brain and the peripheral blood system to maintain homeostasis. Understanding the interactions between infectious agents such as human immunodeficiency virus type 1 (HIV-1), which are capable of traversing the BBB and causing neuroinflammation requires modeling an authentic BBB in vitro. Such an in vitro BBB model also helps develop means of targeting viruses that reside in the brain via natural immune effectors such as antibodies. The BBB consists of human brain microvascular endothelial cells (HBMECs), astrocytes, and pericytes. Here we report in vitro methods to establish a dual-cell BBB model consisting of primary HBMECs and primary astrocytes to measure the integrity of the BBB and antibody penetration of the BBB, as well as a method to establish a single cell BBB model to study the impact of HIV-1 infected medium on the integrity of such a BBB.


Assuntos
Astrócitos , Barreira Hematoencefálica , Células Endoteliais , Infecções por HIV , HIV-1 , Barreira Hematoencefálica/virologia , Barreira Hematoencefálica/metabolismo , Humanos , Astrócitos/virologia , Astrócitos/metabolismo , Astrócitos/imunologia , Células Endoteliais/virologia , Células Endoteliais/metabolismo , Células Endoteliais/imunologia , HIV-1/imunologia , HIV-1/fisiologia , Infecções por HIV/virologia , Infecções por HIV/imunologia , Pericitos/virologia , Pericitos/metabolismo , Pericitos/imunologia , Doenças Neuroinflamatórias/virologia , Doenças Neuroinflamatórias/imunologia , Técnicas de Cocultura/métodos , Células Cultivadas , Encéfalo/virologia , Encéfalo/imunologia , Encéfalo/metabolismo
2.
Int J Mol Sci ; 25(10)2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38791110

RESUMO

Vascular co-option is a consequence of the direct interaction between perivascular cells, known as pericytes (PCs), and glioblastoma multiforme (GBM) cells (GBMcs). This process is essential for inducing changes in the pericytes' anti-tumoral and immunoreactive phenotypes. Starting from the initial stages of carcinogenesis in GBM, PCs conditioned by GBMcs undergo proliferation, acquire a pro-tumoral and immunosuppressive phenotype by expressing and secreting immunosuppressive molecules, and significantly hinder the activation of T cells, thereby facilitating tumor growth. Inhibiting the pericyte (PC) conditioning mechanisms in the GBM tumor microenvironment (TME) results in immunological activation and tumor disappearance. This underscores the pivotal role of PCs as a key cell in the TME, responsible for tumor-induced immunosuppression and enabling GBM cells to evade the immune system. Other cells within the TME, such as tumor-associated macrophages (TAMs) and microglia, have also been identified as contributors to this immunomodulation. In this paper, we will review the role of these three cell types in the immunosuppressive properties of the TME. Our conclusion is that the cellular heterogeneity of immunocompetent cells within the TME may lead to the misinterpretation of cellular lineage identification due to different reactive stages and the identification of PCs as TAMs. Consequently, novel therapies could be developed to disrupt GBM-PC interactions and/or PC conditioning through vascular co-option, thereby exposing GBMcs to the immune system.


Assuntos
Neoplasias Encefálicas , Pericitos , Microambiente Tumoral , Pericitos/imunologia , Pericitos/patologia , Pericitos/metabolismo , Humanos , Microambiente Tumoral/imunologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/metabolismo , Glioma/imunologia , Glioma/patologia , Glioma/metabolismo , Glioblastoma/imunologia , Glioblastoma/patologia , Glioblastoma/metabolismo , Progressão da Doença , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia
3.
J Cell Mol Med ; 26(11): 3203-3212, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35611804

RESUMO

Microvasculature consisting of endothelial cells and pericytes is the main site of injury during antibody-mediated rejection (ABMR) of renal grafts. Little is known about the mechanisms of activation of pericytes in this pathology. We have found recently that activation of Notch3, a mediator of vascular smooth muscle cell proliferation and dedifferentiation, promotes renal inflammation and fibrosis and aggravates progression of renal disease. Therefore, we studied the pericyte expression of Notch3 in 49 non-selected renal graft biopsies (32 for clinical cause, 17 for graft surveillance). We analysed its relationship with patients' clinical and morphological data, and compared with the expression of partial endothelial mesenchymal transition (pEndMT) markers, known to reflect endothelial activation during ABMR. Notch3 was de novo expressed in pericytes of grafts with ABMR, and was significantly correlated with the microcirculation inflammation scores of peritubular capillaritis and glomerulitis and with the expression of pEndMT markers. Notch3 expression was also associated with graft dysfunction and proteinuria at the time of biopsy and in the long term. Multivariate analysis confirmed pericyte expression of Notch3 as an independent risk factor predicting graft loss. These data suggest that Notch3 is activated in the pericytes of renal grafts with ABMR and is associated with poor graft outcome.


Assuntos
Rejeição de Enxerto , Pericitos , Receptor Notch3 , Anticorpos , Biomarcadores/análise , Biópsia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Pericitos/imunologia , Pericitos/patologia , Receptor Notch3/biossíntese , Receptor Notch3/imunologia
4.
Front Immunol ; 12: 704050, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34386010

RESUMO

When combined with anti-PD-1, monoclonal antibodies (mAbs) against GARP:TGF-ß1 complexes induced more frequent immune-mediated rejections of CT26 and MC38 murine tumors than anti-PD-1 alone. In both types of tumors, the activity of anti-GARP:TGF-ß1 mAbs resulted from blocking active TGF-ß1 production and immunosuppression by GARP-expressing regulatory T cells. In CT26 tumors, combined GARP:TGF-ß1/PD-1 blockade did not augment the infiltration of T cells, but did increase the effector functions of already present anti-tumor T cells. Here we show that, in contrast, in MC38, combined GARP:TGF-ß1/PD-1 blockade increased infiltration of T cells, as a result of increased extravasation of T cells from blood vessels. Unexpectedly, combined GARP:TGF-ß1/PD-1 blockade also increased the density of GARP+ blood vessels covered by pericytes in MC38, but not in CT26 tumors. This appears to occur because anti-GARP:TGF-ß1, by blocking TGF-ß1 signals, favors the proliferation of and expression of adhesion molecules such as E-selectin by blood endothelial cells. The resulting densification of intratumoral blood vasculature probably contributes to increased T cell infiltration and to the therapeutic efficacy of GARP:TGF-ß1/PD-1 blockade in MC38. We conclude from these distinct observations in MC38 and CT26, that the combined blockades of GARP:TGF-ß1 and PD-1 can exert anti-tumor activity via multiple mechanisms, including the densification and normalization of intratumoral blood vasculature, the increase of T cell infiltration into the tumor and the increase of the effector functions of intratumoral tumor-specific T cells. This may prove important for the selection of cancer patients who could benefit from combined GARP:TGF-ß1/PD-1 blockade in the clinics.


Assuntos
Antineoplásicos Imunológicos , Vasos Sanguíneos/imunologia , Proteínas de Membrana , Neoplasias Experimentais , Neovascularização Patológica , Pericitos/imunologia , Receptor de Morte Celular Programada 1 , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta1 , Animais , Antineoplásicos Imunológicos/imunologia , Antineoplásicos Imunológicos/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/imunologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/imunologia
5.
J Reprod Immunol ; 145: 103326, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33965695

RESUMO

Decidual stromal cells (DSCs) are the most abundant cellular component of human decidua and play a central role in maternal-fetal immune tolerance. Antigen phenotyping and functional studies recently confirmed the relationship of DSCs with mesenchymal stem/stromal cells (MSCs) and pericytes, the latter two cell types being closely related or identical. The present study investigated the effect of decidualization, a process of cell differentiation driven by progesterone (P4) and other pregnancy hormones, on the MSC/pericyte characteristics of DSCs. To this end we isolated undifferentiated DSC (preDSC) lines that were decidualized in vitro (dDSC) by the effect of P4 and cAMP. Using flow cytometry, we found significant downmodulation of the expression of the MSC/pericyte markers α-smooth muscle actin, nestin, CD140b, CD146 and SUSD2 in dDSCs. The dDSCs did not differ, compared to preDSCs, in the expression of angiogenic factors (characteristic of pericytes) HGF, FGF2, ANGPT1 or VEGF according to RT-PCR results, but had significantly increased PGF expression. In migration assays, preDSC-conditioned media had a chemotactic effect on the THP-1 monocytic line (characteristic of pericytes), and this effect was significantly greater in dDSC-conditioned media. Media conditioned with dDSC, but not with preDSC, induced apoptosis in 4 out of 6 different tumor cell lines (characteristic of MSCs) according to propidium iodide staining and flow cytometry results. Our findings show that decidualization induces phenotypic and functional changes in the MSC/pericyte properties of DSCs that may have a role in the normal development of pregnancy.


Assuntos
Decídua/crescimento & desenvolvimento , Histocompatibilidade Materno-Fetal , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Neoplasias/terapia , Adulto , Antígenos/metabolismo , Diferenciação Celular/imunologia , Fatores Quimiotáticos/metabolismo , Quimiotaxia/imunologia , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Decídua/citologia , Decídua/imunologia , Feminino , Voluntários Saudáveis , Humanos , Células-Tronco Mesenquimais/metabolismo , Neoplasias/imunologia , Pericitos/imunologia , Pericitos/metabolismo , Gravidez , Células THP-1 , Adulto Jovem
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33653955

RESUMO

Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood-brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret ), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti-ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret ;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.


Assuntos
Barreira Hematoencefálica/imunologia , Encefalomielite Autoimune Experimental/imunologia , Homeostase/imunologia , Leucócitos/imunologia , Pericitos/imunologia , Animais , Barreira Hematoencefálica/patologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Homeostase/genética , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Leucócitos/patologia , Camundongos , Camundongos Transgênicos , Pericitos/patologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
7.
J Neurosci Res ; 98(12): 2390-2405, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32815569

RESUMO

Pericytes are contractile cells that extend along the vasculature to mediate key homeostatic functions of endothelial barriers within the body. In the central nervous system (CNS), pericytes are important contributors to the structure and function of the neurovascular unit, which includes endothelial cells, astrocytes and neurons. The understanding of pericytes has been marred by an inability to accurately distinguish pericytes from other stromal cells with similar expression of identifying markers. Evidence is now growing in favor of pericytes being actively involved in both CNS homeostasis and pathology of neurological diseases, including multiple sclerosis, spinal cord injury, and Alzheimer's disease among others. In this review, we discuss the current understanding on the characterization of pericytes, their roles in maintaining the integrity of the blood-brain barrier, and their contributions to neuroinflammation and neurorepair. Owing to its plethora of surface receptors, pericytes respond to inflammatory mediators such as CCL2 (monocyte chemoattractant protein-1) and tumor necrosis factor-α, in turn secreting CCL2, nitric oxide, and several cytokines. Pericytes can therefore act as promoters of both the innate and adaptive arms of the immune system. Much like professional phagocytes, pericytes also have the ability to clear up cellular debris and macromolecular plaques. Moreover, pericytes promote the activities of CNS glia, including in maturation of oligodendrocyte lineage cells for myelination. Conversely, pericytes can impair regenerative processes by contributing to scar formation. A better characterization of CNS pericytes and their functions would bode well for therapeutics aimed at alleviating their undesirable properties and enhancing their benefits.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Homeostase/fisiologia , Mediadores da Inflamação/metabolismo , Doenças do Sistema Nervoso/metabolismo , Pericitos/metabolismo , Animais , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Endocitose/fisiologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Mediadores da Inflamação/imunologia , Doenças do Sistema Nervoso/imunologia , Pericitos/imunologia
8.
Cancer Immunol Res ; 8(8): 990-1003, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32580941

RESUMO

Metastasis of human tumors to lymph nodes (LN) is a universally negative prognostic factor. LN stromal cells (SC) play a crucial role in enabling T-cell responses, and because tumor metastases modulate their structure and function, this interaction may suppress immune responses to tumor antigens. The SC subpopulations that respond to infiltration of malignant cells into human LNs have not been defined. Here, we identify distinctive subpopulations of CD90+ SCs present in melanoma-infiltrated LNs and compare them with their counterparts in normal LNs. The first population (CD90+ podoplanin+ CD105+ CD146+ CD271+ VCAM-1+ ICAM-1+ α-SMA+) corresponds to fibroblastic reticular cells that express various T-cell modulating cytokines, chemokines, and adhesion molecules. The second (CD90+ CD34+ CD105+ CD271+) represents a novel population of CD34+ SCs embedded in collagenous structures, such as the capsule and trabeculae, that predominantly produce extracellular matrix. We also demonstrated that these two SC subpopulations are distinct from two subsets of human LN pericytes, CD90+ CD146+ CD36+ NG2- pericytes in the walls of high endothelial venules and other small vessels, and CD90+ CD146+ NG2+ CD36- pericytes in the walls of larger vessels. Distinguishing between these CD90+ SC subpopulations in human LNs allows for further study of their respective impact on T-cell responses to tumor antigens and clinical outcomes.


Assuntos
Biomarcadores Tumorais/imunologia , Linfonodos/imunologia , Melanoma/imunologia , Pericitos/imunologia , Células Estromais/imunologia , Biomarcadores Tumorais/metabolismo , Diferenciação Celular/imunologia , Humanos , Imunofenotipagem/métodos , Linfonodos/patologia , Melanoma/classificação , Melanoma/patologia , Metástase Neoplásica , Pericitos/patologia , Células Estromais/patologia , Evasão Tumoral
9.
Mol Biol Rep ; 47(5): 3833-3856, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32361895

RESUMO

In this study we used two different techniques in order to isolate pericytes from the wall of human umbilical cord vein and get two different groups of cells were named as "pellet and primer cells". These groups were compared with each other according to their morphologies and stem cell marker expressions. Also, these two different populations were compared with each other and with human bone marrow mesenchymal stem cells (BM-MSCs) according to their transcriptomic profiles. Then, pellet cells proteomic profiles were determined. Our results showed that morphologies and cell surface marker expressions of pellet cells and primer cells are similar. On the other hand, according to immunofluorescence staining results, in contrast to primer cells, pellet cells showed positive NG2 and PDGFR-ß staining. As a result of gene expression profiling, pellet cells have upregulated genes related with muscle, neural and immune cell differentiation, development and pluripotency. On the other hand, primer cells have upregulated adhesion pathway-related genes. In addition to differences between pellet and primer cells, the gene expression profiles of these cell groups are also different from BM-MSCs. The results of transcriptome and proteome analysis of pellet cells were in consistent with each other.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Pericitos/citologia , Veias Umbilicais/citologia , Adulto , Células da Medula Óssea/citologia , Antígeno CD146/biossíntese , Antígeno CD146/imunologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Sangue Fetal/citologia , Sangue Fetal/imunologia , Sangue Fetal/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica/métodos , Células-Tronco Embrionárias Humanas/imunologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Pericitos/imunologia , Pericitos/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Células-Tronco/metabolismo , Transcriptoma , Cordão Umbilical/citologia , Veias Umbilicais/metabolismo
10.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L813-L830, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32073879

RESUMO

Our understanding of mesenchymal cell subsets and their function in human lung affected by aging and in certain disease settings remains poorly described. We use a combination of flow cytometry, prospective cell-sorting strategies, confocal imaging, and modeling of microvessel formation using advanced microfluidic chip technology to characterize mesenchymal cell subtypes in human postnatal and adult lung. Tissue was obtained from patients undergoing elective surgery for congenital pulmonary airway malformations (CPAM) and other airway abnormalities including chronic obstructive pulmonary disease (COPD). In microscopically normal postnatal human lung, there was a fivefold higher mesenchymal compared with epithelial (EpCAM+) fraction, which diminished with age. The mesenchymal fraction composed of CD90+ and CD90+CD73+ cells was enriched in CXCL12 and platelet-derived growth factor receptor-α (PDGFRα) and located in close proximity to EpCAM+ cells in the alveolar region. Surprisingly, alveolar organoids generated from EpCAM+ cells supported by CD90+ subset were immature and displayed dysplastic features. In congenital lung lesions, cystic air spaces and dysplastic alveolar regions were marked with an underlying thick interstitium composed of CD90+ and CD90+PDGFRα+ cells. In postnatal lung, a subset of CD90+ cells coexpresses the pericyte marker CD146 and supports self-assembly of perfusable microvessels. CD90+CD146+ cells from COPD patients fail to support microvessel formation due to fibrinolysis. Targeting the plasmin-plasminogen system during microvessel self-assembly prevented fibrin gel degradation, but microvessels were narrower and excessive contraction blocked perfusion. These data provide important new information regarding the immunophenotypic identity of key mesenchymal lineages and their change in a diverse setting of congenital lung lesions and COPD.


Assuntos
Imunomodulação/imunologia , Células-Tronco Mesenquimais/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo , Adolescente , Biomarcadores/metabolismo , Antígeno CD146/imunologia , Antígeno CD146/metabolismo , Separação Celular/métodos , Criança , Pré-Escolar , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Feminino , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Lactente , Recém-Nascido , Masculino , Células-Tronco Mesenquimais/imunologia , Microvasos/imunologia , Microvasos/metabolismo , Pericitos/imunologia , Pericitos/metabolismo , Estudos Prospectivos
11.
Proc Natl Acad Sci U S A ; 116(41): 20655-20665, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31548426

RESUMO

The contractile perivascular cells, pericytes (PC), are hijacked by glioblastoma (GB) to facilitate tumor progression. PC's protumorigenic function requires direct interaction with tumor cells and contributes to the establishment of immunotolerance to tumor growth. Cancer cells up-regulate their own chaperone-mediated autophagy (CMA), a process that delivers selective cytosolic proteins to lysosomes for degradation, with pro-oncogenic effects. However, the possible impact that cancer cells may have on CMA of surrounding host cells has not been explored. We analyzed the contribution of CMA to the GB-induced changes in PC biology. We have found that CMA is markedly up-regulated in PC in response to the oxidative burst that follows PC-GB cell interaction. Genetic manipulations to block the GB-induced up-regulation of CMA in PC allows them to maintain their proinflammatory function and to support the induction of effective antitumor T cell responses required for GB clearance. GB-induced up-regulation of CMA activity in PC is essential for their effective interaction with GB cells that help tumor growth. We show that CMA inhibition in PC promotes GB cell death and the release of high immunogenic levels of granulocyte-macrophage colony stimulating factor (GM-CSF), through deregulation of the expression of cell-to-cell interaction proteins and protein secretion. A GB mouse model grafted in vivo with CMA-defective PC shows reduced GB proliferation and effective immune response compared to mice grafted with control PC. Our findings identify abnormal up-regulation of CMA as a mechanism by which GB cells elicit the immunosuppressive function of PC and stabilize GB-PC interactions necessary for tumor cell survival.


Assuntos
Apoptose , Autofagia Mediada por Chaperonas , Glioblastoma/patologia , Chaperonas Moleculares/metabolismo , Pericitos/imunologia , Animais , Proliferação de Células , Glioblastoma/imunologia , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Pericitos/metabolismo , Pericitos/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Sci Rep ; 9(1): 9745, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31278356

RESUMO

Prophylactic intracameral injection of antibiotics is commonly used to prevent endophthalmitis after cataract surgery. However, devastating visual complications have been reported including hemorrhagic occlusive retinal vasculitis (HORV).To determine the toxic and inflammatory effects of moxifloxacin, cefuroxime, and vancomycin on human retinal vascular cells, human retinal vascular endothelial cells (RVEC) and pericytes were exposed to three antibiotics, and the adverse effects were assessed by membrane damage, loss of intrinsic esterase activity, kinetic cell viability, and inflammatory cytokine secretion. Their retinal toxicity was examined by live/dead assays after an intravitreal injection of the three antibiotics into mice eyes. In vascular cells in culture, membrane damage and loss of esterase activity were induced after exposure to the three antibiotics. The toxic effects were most obvious after moxifloxacin (RVEC, ≥125 µg/mL; pericytes, ≥1000 µg/mL) at 24 h. Cefuroxime also reduced esterase activity and the membrane integrity of vascular cells but were less toxic than moxifloxacin. Kinetic cell viability testing showed that 500 µg/mL of moxifloxacin exposure induced significant decrease (29%) in the viability as early as 1 h. When the inflammatory effects of the antibiotics were examined, a significant induction of IL-8 was observed especially by RVECs after exposure to cefuroxime or vancomycin which was exacerbated by L-alanyl-γ-D-glutamyl-meso-diaminopimelic acid (Tri-DAP), a NOD1 ligand. Intravitreal injections in mice showed that cefuroxime and vancomycin caused retinal and vascular toxicity extending to the inner nuclear layers. Collectively, moxifloxacin causes immediate damage to retinal vascular cells in vitro, while cefuroxime and vancomycin induced significant inflammatory effects on vascular endothelial cells and caused retinal toxicity. Surgeons need to be cautious of the toxicity when antibiotics are used prophylactically especially by intravitreal administration.


Assuntos
Antibacterianos/efeitos adversos , Interleucina-8/metabolismo , Pericitos/citologia , Retina/citologia , Animais , Antibacterianos/administração & dosagem , Cefuroxima/administração & dosagem , Cefuroxima/efeitos adversos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Esterases/metabolismo , Humanos , Injeções Intravítreas , Camundongos , Moxifloxacina/administração & dosagem , Moxifloxacina/efeitos adversos , Pericitos/efeitos dos fármacos , Pericitos/imunologia , Retina/efeitos dos fármacos , Retina/imunologia , Vancomicina/administração & dosagem , Vancomicina/efeitos adversos
13.
Mediators Inflamm ; 2019: 4123605, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31205449

RESUMO

Leukocyte recruitment is a hallmark of the inflammatory response. Migrating leukocytes breach the endothelium along with the vascular basement membrane and associated pericytes. While much is known about leukocyte-endothelial cell interactions, the mechanisms and role of pericytes in extravasation are poorly understood and the classical paradigm of leukocyte recruitment in the microvasculature seldom adequately discusses the involvement of pericytes. Emerging evidence shows that pericytes are essential players in the regulation of leukocyte extravasation in addition to their functions in blood vessel formation and blood-brain barrier maintenance. Junctions between venular endothelial cells are closely aligned with extracellular matrix protein low expression regions (LERs) in the basement membrane, which in turn are aligned with gaps between pericytes. This forms preferential paths for leukocyte extravasation. Breaching of the layer formed by pericytes and the basement membrane entails remodelling of LERs, leukocyte-pericyte adhesion, crawling of leukocytes on pericyte processes, and enlargement of gaps between pericytes to form channels for migrating leukocytes. Furthermore, inflamed arteriolar and capillary pericytes induce chemotactic migration of leukocytes that exit postcapillary venules, and through direct pericyte-leukocyte contact, they induce efficient interstitial migration to enhance the immunosurveillance capacity of leukocytes. Given their role as regulators of leukocyte extravasation, proper pericyte function is imperative in inflammatory disease contexts such as diabetic retinopathy and sepsis. This review summarizes research on the molecular mechanisms by which pericytes mediate leukocyte diapedesis in inflamed tissues.


Assuntos
Leucócitos/metabolismo , Pericitos/metabolismo , Animais , Membrana Basal/imunologia , Membrana Basal/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Leucócitos/imunologia , Pericitos/imunologia
14.
Genes (Basel) ; 10(6)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31234442

RESUMO

Osteoarthritis (OA) is a degenerative joint disease accompanied by pain and loss of function. Adipose tissue harbors mesenchymal stem/stromal cells (MSC), or medicinal signaling cells as suggested by Caplan (Caplan, 2017), used in autologous transplantation in many clinical settings. The aim of the study was to characterize a stromal vascular fraction from microfragmented lipoaspirate (SVF-MLA) applied for cartilage treatment in OA and compare it to that of autologous lipoaspirate (SVF-LA). Samples were first stained using a DuraClone SC prototype tube for the surface detection of CD31, CD34, CD45, CD73, CD90, CD105, CD146 and LIVE/DEAD Yellow Fixable Stain for dead cell detection, followed by DRAQ7 cell nuclear dye staining, and analyzed by flow cytometry. In SVF-LA and SVF-MLA samples, the following population phenotypes were identified within the CD45- fraction: CD31+CD34+CD73±CD90±CD105±CD146± endothelial progenitors (EP), CD31+CD34-CD73±CD90±CD105-CD146± mature endothelial cells, CD31-CD34-CD73±CD90+CD105-CD146+ pericytes, CD31-CD34+CD73±CD90+CD105-CD146+ transitional pericytes, and CD31-CD34+CD73highCD90+CD105-CD146- supra-adventitial-adipose stromal cells (SA-ASC). The immunophenotyping profile of SVF-MLA was dominated by a reduction of leukocytes and SA-ASC, and an increase in EP, evidencing a marked enrichment of this cell population in the course of adipose tissue microfragmentation. The role of EP in pericyte-primed MSC-mediated tissue healing, as well as the observed hormonal implication, is yet to be investigated.


Assuntos
Túnica Adventícia/imunologia , Cartilagem/metabolismo , Imunofenotipagem , Osteoartrite/tratamento farmacológico , Adipócitos/efeitos dos fármacos , Adipócitos/imunologia , Túnica Adventícia/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Feminino , Citometria de Fluxo , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Osteoartrite/imunologia , Osteoartrite/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/imunologia
15.
Front Immunol ; 10: 266, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30886614

RESUMO

Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of synovium (synovitis), with inflammatory/immune cells and resident fibroblast-like synoviocytes (FLS) acting as major players in the pathogenesis of this disease. The resulting inflammatory response poses considerable risks as loss of bone and cartilage progresses, destroying the joint surface, causing joint damage, joint failure, articular dysfunction, and pre-mature death if left untreated. At the cellular level, early changes in RA synovium include inflammatory cell infiltration, synovial hyperplasia, and stimulation of angiogenesis to the site of injury. Different angiogenic factors promote this disease, making the role of anti-angiogenic therapy a focus of RA treatment. To control angiogenesis, mesenchymal stromal cells/pericytes (MSCs) in synovial tissue play a vital role in tissue repair. While recent evidence reports that MSCs found in joint tissues can differentiate to repair damaged tissue, this repair function can be repressed by the inflammatory milieu. Extremely-low frequency pulsed electromagnetic field (PEMF), a biophysical form of stimulation, has an anti-inflammatory effect by causing differentiation of MSCs. PEMF has also been reported to increase the functional activity of MSCs to improve differentiation to chondrocytes and osteocytes. Moreover, PEMF has been demonstrated to accelerate cell differentiation, increase deposition of collagen, and potentially return vascular dysfunction back to homeostasis. The aim of this report is to review the effects of PEMF on MSC modulation of cytokines, growth factors, and angiogenesis, and describe its effect on MSC regeneration of synovial tissue to further understand its potential role in the treatment of RA.


Assuntos
Artrite Reumatoide/imunologia , Células-Tronco Mesenquimais/imunologia , Pericitos/imunologia , Animais , Diferenciação Celular/imunologia , Citocinas/imunologia , Campos Eletromagnéticos , Humanos , Inflamação/imunologia
16.
J Cell Biol ; 218(2): 700-721, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30626719

RESUMO

Cocaine is known to facilitate the transmigration of inflammatory leukocytes into the brain, an important mechanism underlying neuroinflammation. Pericytes are well-recognized as important constituents of the blood-brain barrier (BBB), playing a key role in maintaining barrier integrity. In the present study, we demonstrate for the first time that exposure of human brain vascular pericytes to cocaine results in enhanced secretion of CXCL10, leading, in turn, to increased monocyte transmigration across the BBB both in vitro and in vivo. This process involved translocation of σ-1 receptor (σ-1R) and interaction of σ-1R with c-Src kinase, leading to activation of the Src-PDGFR-ß-NF-κB pathway. These findings imply a novel role for pericytes as a source of CXCL10 in the pericyte-monocyte cross talk in cocaine-mediated neuroinflammation, underpinning their role as active components of the innate immune responses.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/toxicidade , Quimiocina CXCL10/metabolismo , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Cocaína/toxicidade , Monócitos/efeitos dos fármacos , Comunicação Parácrina/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Proteína Tirosina Quinase CSK/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Estudos de Casos e Controles , Transtornos Relacionados ao Uso de Cocaína/imunologia , Técnicas de Cocultura , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/imunologia , Monócitos/metabolismo , NF-kappa B/metabolismo , Pericitos/imunologia , Pericitos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores sigma/metabolismo , Transdução de Sinais , Receptor Sigma-1
17.
J Vis Exp ; (140)2018 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-30346386

RESUMO

Retinal pericytes play an important role in many diseases of the eye. Immunohistochemical staining techniques of retinal vessels and microvascular pericytes are central to ophthalmological research. It is vital to choose an appropriate method of visualizing the microvascular pericytes. We describe retinal microvascular pericyte immunohistochemical staining in cryo-sections, whole-mounts, and hypotonic isolated vasculature using antibodies for platelet-derived growth factor receptor ß (PDGFRß) and nerve/glial antigen 2 (NG2). This allows us to highlight advantages and shortcomings of each of the three tissue preparations for the visualization of the retinal microvascular pericytes. Cryo-sections provide transsectional visualization of all retinal layers but contain only a few occasional transverse cuts of the microvasculature. Whole-mount provides an overview of the entire retinal vasculature, but visualization of the microvasculature can be troublesome. Hypotonic isolation provides a method to visualize the entire retinal vasculature by the removal of neuronal cells, but this makes the tissue very fragile.


Assuntos
Técnicas Histológicas/métodos , Imuno-Histoquímica/métodos , Pericitos/imunologia , Retina/diagnóstico por imagem , Vasos Retinianos/imunologia , Animais , Humanos , Ratos , Retina/metabolismo
18.
Int J Mol Sci ; 19(10)2018 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-30257508

RESUMO

Successful vascularization is essential in wound healing, the histo-integration of biomaterials, and other aspects of regenerative medicine. We developed a functional in vitro assay to dissect the complex processes directing angiogenesis during wound healing, whereby vascular cell spheroids were induced to sprout in the presence of classically (M1) or alternatively (M2) activated macrophages. This simulated a microenvironment, in which sprouting cells were exposed to the inflammatory or proliferation phases of wound healing, respectively. We showed that M1 macrophages induced single-cell migration of endothelial cells and pericytes. In contrast, M2 macrophages augmented endothelial sprouting, suggesting that vascular cells infiltrate the wound bed during the inflammatory phase and extensive angiogenesis is initiated upon a switch to a predominance of M2. Interestingly, M1 and M2 shared a pro-angiogenic secretome, whereas pro-inflammatory cytokines were solely secreted by M1. These results suggested that acute inflammatory factors act as key inducers of vascular cell infiltration and as key negative regulators of angiogenesis, whereas pro-angiogenic factors are present throughout early wound healing. This points to inflammatory factors as key targets to modulate angiogenesis. The here-established wound healing assay represents a useful tool to investigate the effect of biomaterials and factors on angiogenesis during wound healing.


Assuntos
Proliferação de Células , Inflamação/imunologia , Ativação de Macrófagos , Neovascularização Fisiológica , Cicatrização , Linhagem Celular , Movimento Celular , Citocinas/imunologia , Células Endoteliais/citologia , Células Endoteliais/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Mediadores da Inflamação/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Pericitos/citologia , Pericitos/imunologia
19.
Mol Vis ; 24: 379-394, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29853772

RESUMO

Purpose: The purpose of this study was to determine whether the blood-retina barrier is compromised by choroidal murine cytomegalovirus (MCMV) infection, using electron microscopy. Methods: BALB/c mice were immunosuppressed with methylprednisolone and monoclonal antibodies to CD4 and CD8. At several time points post-MCMV intraperitoneal inoculation, the eyes were removed and analyzed with western blotting and immunoelectron microscopy for the presence of MCMV early antigen (EA) and the host protein RIP3. Posterior eyecups from RIP3-/- and RIP3+/+ mice were cultured and inoculated with MCMV. At days 4, 7, and 11 post-infection, cultures were collected and analyzed with plaque assay, immunohistochemical staining, and real-time PCR (RT-PCR). Results: MCMV EA was observed in the nuclei of vascular endothelial cells and pericytes in the choriocapillaris. Disruption of Bruch's membrane was observed, especially at sites adjacent to activated platelets, and a few RPE cells containing some enlarged vesicles were found directly beneath disrupted Bruch's membrane. Some virus particles were also observed in the enlarged vesicles of RPE cells. Levels of the RIP3 protein, which was observed mainly in the RPE cells and the basement membrane of the choriocapillaris, were greatly increased following MCMV infection, while depletion of RIP3 resulted in greatly decreased inflammasome formation, as well as expression of downstream inflammation factors. Conclusions: The results suggest that systemic MCMV spreads to the choroid and replicates in vascular endothelia and pericytes of the choriocapillaris during immunosuppression. Choroidal MCMV infection is associated with in situ inflammation and subsequent disruption of Bruch's membrane and the outer blood-retina barrier.


Assuntos
Corioide/imunologia , Infecções por Citomegalovirus/imunologia , Infecções Oculares Virais/imunologia , Hospedeiro Imunocomprometido , Retina/imunologia , Retinite/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos Virais/genética , Plaquetas/imunologia , Plaquetas/patologia , Plaquetas/virologia , Barreira Hematorretiniana/imunologia , Barreira Hematorretiniana/patologia , Barreira Hematorretiniana/virologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Corioide/irrigação sanguínea , Corioide/patologia , Corioide/virologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Células Endoteliais , Infecções Oculares Virais/patologia , Infecções Oculares Virais/virologia , Feminino , Proteínas Imediatamente Precoces/genética , Inflamassomos/imunologia , Metilprednisolona/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/crescimento & desenvolvimento , Muromegalovirus/patogenicidade , Pericitos/imunologia , Pericitos/patologia , Pericitos/virologia , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Retina/patologia , Retina/virologia , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/virologia , Retinite/patologia , Retinite/virologia
20.
Cell Transplant ; 27(12): 1798-1808, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29845887

RESUMO

Stroke is a cerebrovascular disorder that affects many people worldwide. In addition to the well-established functions of astrocytes and microglia in stroke pathogenesis, pericytes also play an important role in stroke progression and recovery. As perivascular multi-potent cells and an important component of the blood-brain barrier (BBB), pericytes have been shown to exert a large variety of functions, including serving as stem/progenitor cells and maintaining BBB integrity. Here in this review, we summarize the roles of pericytes in stroke pathogenesis, with a focus on their effects in cerebral blood flow, BBB integrity, angiogenesis, immune responses, scar formation and fibrosis.


Assuntos
Barreira Hematoencefálica/metabolismo , Pericitos/metabolismo , Acidente Vascular Cerebral/patologia , Cicatriz/patologia , Humanos , Neovascularização Patológica , Pericitos/imunologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA