Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 587
Filtrar
1.
J Biophotonics ; 16(4): e202200310, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36519190

RESUMO

Psoriasis is a chronic inflammatory skin disease involved with both complex morphological changes of skin and immune processes. The clinical diagnostics and research of psoriasis often require invasive biopsy which lacks their real-time dynamics in vivo. Here we report a noninvasive microscopic system developed by combining in vivo fluorescent microscopy, optical clearing, and immunolabeling to enable real-time imaging of immune cells and cytokines in blood flow in psoriatic animal models. The vascular morphology and time-lapse kinetics of interleukin (IL)-23, IL-17, tumor necrosis factor-α, and CD4+ cells in blood are captured at submicron resolution through the thickening epidermis and opaque scales during the development of psoriasis in vivo. Our data suggest IL-23 recruits CD4+ cells to release IL-17 in blood that further leaks out in the psoriatic skin area. This optical system enables noninvasive and real-time assessment of immune molecules and cells in vivo, providing good potential for medical researches on psoriasis.


Assuntos
Microscopia de Fluorescência , Imagem Óptica , Psoríase , Pele , Animais , Camundongos , Psoríase/sangue , Psoríase/diagnóstico por imagem , Psoríase/imunologia , Modelos Animais de Doenças , Microscopia de Fluorescência/métodos , Interleucina-23/sangue , Interleucina-17/sangue , Fator de Necrose Tumoral alfa/sangue , Linfócitos T CD4-Positivos/imunologia , Pele/diagnóstico por imagem , Pele/imunologia , Imagem Óptica/métodos , Vasos Sanguíneos/diagnóstico por imagem , Vasos Sanguíneos/imunologia
2.
Biomolecules ; 12(12)2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36551229

RESUMO

Interleukin-18 (IL-18) can effectively activate natural killer (NK) cells and induce large concentrations of interferon-γ (IFN-γ). In healthy humans, IL-18 binding protein (IL-18BP) can inhibit the binding of IL-18 to IL-18R and counteract the biological action of IL-18 due to its high concentration and high affinity, thus preventing the production of IFN-γ and inhibiting NK-cell activation. Through previous studies and the phenomena observed by our group in pig-non-human primates (NHPs) liver transplantation experiments, we proposed that the imbalance in IL-18/IL-18BP expression upon transplantation encourages the activation, proliferation, and cytotoxic effects of NK cells, ultimately causing acute vascular rejection of the graft. In this research, we used Lewis-Brown Norway rat orthotopic liver transplantation (OLTx) as a model of acute vascular rejection. AAV8-Il18bp viral vectors as gene delivery vehicles were constructed for gene therapy to overexpress IL-18BP and alleviate NK-cell rejection of the graft after transplantation. The results showed that livers overexpressing IL-18BP had reduced damage and could function longer after transplantation, effectively improving the survival time of the recipients.


Assuntos
Vasos Sanguíneos , Terapia Genética , Rejeição de Enxerto , Sobrevivência de Enxerto , Interleucina-18 , Transplante de Fígado , Animais , Ratos , Rejeição de Enxerto/prevenção & controle , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Transplante de Fígado/métodos , Ratos Endogâmicos Lew , Sobrevivência de Enxerto/genética , Vasos Sanguíneos/imunologia , Vetores Genéticos
3.
Respir Res ; 23(1): 25, 2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-35144622

RESUMO

BACKGROUND: Pulmonary hyperinflammation is a key event with SARS-CoV-2 infection. Acute respiratory distress syndrome (ARDS) that often accompanies COVID-19 appears to have worse outcomes than ARDS from other causes. To date, numerous lung histological studies in cases of COVID-19 have shown extensive inflammation and injury, but the extent to which these are a COVID-19 specific, or are an ARDS and/or mechanical ventilation (MV) related phenomenon is not clear. Furthermore, while lung hyperinflammation with ARDS (COVID-19 or from other causes) has been well studied, there is scarce documentation of vascular inflammation in COVID-19 lungs. METHODS: Lung sections from 8 COVID-19 affected and 11 non-COVID-19 subjects, of which 8 were acute respiratory disease syndrome (ARDS) affected (non-COVID-19 ARDS) and 3 were from subjects with non-respiratory diseases (non-COVID-19 non-ARDS) were H&E stained to ascertain histopathological features. Inflammation along the vessel wall was also monitored by expression of NLRP3 and caspase 1. RESULTS: In lungs from COVID-19 affected subjects, vascular changes in the form of microthrombi in small vessels, arterial thrombosis, and organization were extensive as compared to lungs from non-COVID-19 (i.e., non-COVID-19 ARDS and non-COVID-19 non-ARDS) affected subjects. The expression of NLRP3 pathway components was higher in lungs from COVID-19 ARDS subjects as compared to non-COVID-19 non-ARDS cases. No differences were observed between COVID-19 ARDS and non-COVID-19 ARDS lungs. CONCLUSION: Vascular changes as well as NLRP3 inflammasome pathway activation were not different between COVID-19 and non-COVID-19 ARDS suggesting that these responses are not a COVID-19 specific phenomenon and are possibly more related to respiratory distress and associated strategies (such as MV) for treatment.


Assuntos
Vasos Sanguíneos/imunologia , COVID-19/imunologia , Inflamassomos/análise , Pulmão/irrigação sanguínea , Proteína 3 que Contém Domínio de Pirina da Família NLR/análise , Idoso , Idoso de 80 Anos ou mais , Autopsia , Vasos Sanguíneos/patologia , COVID-19/mortalidade , COVID-19/patologia , COVID-19/virologia , Estudos de Casos e Controles , Feminino , Imunofluorescência , Humanos , Masculino , Pessoa de Meia-Idade
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.
Am J Physiol Gastrointest Liver Physiol ; 321(2): G200-G212, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34105986

RESUMO

Hepatosplanchnic and pulmonary vasculatures constitute synapomorphic, highly comparable networks integrated with the external environment. Given functionality related to obligatory requirements of "feeding and breathing," these organs are subject to constant environmental challenges entailing infectious risk, antigenic and xenobiotic exposures. Host responses to these stimuli need to be both protective and tightly regulated. These functions are facilitated by dualistic, high-low pressure blood supply of the liver and lungs, as well as tolerogenic characteristics of resident immune cells and signaling pathways. Dysregulation in hepatosplanchnic and pulmonary blood flow, immune responses, and microbiome implicate common pathogenic mechanisms across these vascular networks. Hepatosplanchnic diseases, such as cirrhosis and portal hypertension, often impact lungs and perturb pulmonary circulation and oxygenation. The reverse situation is also noted with lung disease resulting in hepatic dysfunction. Others, and we, have described common features of dysregulated cell signaling during liver and lung inflammation involving extracellular purines (e.g., ATP, ADP), either generated exogenously or endogenously. These metabokines serve as danger signals, when released by bacteria or during cellular stress and cause proinflammatory and prothrombotic signals in the gut/liver-lung vasculature. Dampening of these danger signals and organ protection largely depends upon activities of vascular and immune cell-expressed ectonucleotidases (CD39 and CD73), which convert ATP and ADP into anti-inflammatory adenosine. However, in many inflammatory disorders involving gut, liver, and lung, these protective mechanisms are compromised, causing perpetuation of tissue injury. We propose that interventions that specifically target aberrant purinergic signaling might prevent and/or ameliorate inflammatory disorders of the gut/liver and lung axis.


Assuntos
Vasos Sanguíneos/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Receptores Purinérgicos/metabolismo , Sepse/metabolismo , Transdução de Sinais , Animais , Vasos Sanguíneos/imunologia , Humanos , Fígado/irrigação sanguínea , Pulmão/irrigação sanguínea , Nucleotídeos de Purina/metabolismo
6.
Int J Mol Sci ; 22(8)2021 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-33920100

RESUMO

Chitinase 3-like 1 (CHI3L1) is an enzymatically inactive mammalian chitinase that is associated with tumor inflammation. Previous research indicated that CHI3L1 is able to interact with different extracellular matrix components, such as heparan sulfate. In the present work, we investigated whether the interaction of CHI3L1 with the extracellular matrix of melanoma cells can trigger an inflammatory activation of endothelial cells. The analysis of the melanoma cell secretome indicated that CHI3L1 increases the abundance of various cytokines, such as CC-chemokine ligand 2 (CCL2), and growth factors, such as vascular endothelial growth factor A (VEGF-A). Using a solid-phase binding assay, we found that heparan sulfate-bound VEGF-A and CCL2 were displaced by recombinant CHI3L1 in a dose-dependent manner. Microfluidic experiments indicated that the CHI3L1 altered melanoma cell secretome promoted immune cell recruitment to the vascular endothelium. In line with the elevated VEGF-A levels, CHI3L1 was also able to promote angiogenesis through the release of extracellular matrix-bound pro-angiogenic factors. In conclusion, we showed that CHI3L1 is able to affect the tumor cell secretome, which in turn can regulate immune cell recruitment and blood vessel formation. Accordingly, our data suggest that the molecular targeting of CHI3L1 in the course of cancer immunotherapies can tune patients' response and antitumoral inflammation.


Assuntos
Quimiocina CCL2/genética , Proteína 1 Semelhante à Quitinase-3/genética , Melanoma/genética , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Linhagem Celular Tumoral , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/crescimento & desenvolvimento , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Matriz Extracelular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicosaminoglicanos/farmacologia , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Melanoma/imunologia , Melanoma/patologia , Técnicas Analíticas Microfluídicas , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Ligação Proteica/genética , Ligação Proteica/imunologia
7.
Front Immunol ; 12: 667830, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33897716

RESUMO

Macrophages are critical mediators of tissue vascularization both in health and disease. In multiple tissues, macrophages have been identified as important regulators of both blood and lymphatic vessel growth, specifically following tissue injury and in pathological inflammatory responses. In development, macrophages have also been implicated in limiting vascular growth. Hence, macrophages provide an important therapeutic target to modulate tissue vascularization in the clinic. However, the molecular mechanisms how macrophages mediate tissue vascularization are still not entirely resolved. Furthermore, mechanisms might also vary among different tissues. Here we review the role of macrophages in tissue vascularization with a focus on their role in blood and lymphatic vessel formation in the barrier tissues cornea and skin. Comparing mechanisms of macrophage-mediated hem- and lymphangiogenesis in the angiogenically privileged cornea and the physiologically vascularized skin provides an opportunity to highlight similarities but also tissue-specific differences, and to understand how macrophage-mediated hem- and lymphangiogenesis can be exploited for the treatment of disease, including corneal wound healing after injury, graft rejection after corneal transplantation or pathological vascularization of the skin.


Assuntos
Vasos Sanguíneos/metabolismo , Córnea/irrigação sanguínea , Neovascularização da Córnea , Linfangiogênese , Vasos Linfáticos/metabolismo , Macrófagos/metabolismo , Neovascularização Fisiológica , Pele/irrigação sanguínea , Animais , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Humanos , Vasos Linfáticos/imunologia , Vasos Linfáticos/patologia , Macrófagos/imunologia , Macrófagos/patologia , Fenótipo , Transdução de Sinais , Cicatrização
8.
Sci Rep ; 11(1): 3614, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33574432

RESUMO

Atherosclerosis is characterized by retention of modified lipoproteins, especially oxidized low density lipoprotein (oxLDL) within the sub-endothelial space of affected blood vessels. Recruited monocyte-derived and tissue-resident macrophages subsequently ingest oxLDL by binding and internalizing oxLDL via scavenger receptors, particularly CD36. The secreted neurorepellent, Slit2, acting through its transmembrane receptor, Roundabout-1 (Robo-1), was previously shown to inhibit recruitment of monocytes into nascent atherosclerotic lesions. The effects of Slit2 on oxLDL uptake by macrophages have not been explored. We report here that Slit2 inhibits uptake of oxLDL by human and murine macrophages, and the resulting formation of foam cells, in a Rac1-dependent and CD36-dependent manner. Exposure of macrophages to Slit2 prevented binding of oxLDL to the surface of cells. Using super-resolution microscopy, we observed that exposure of macrophages to Slit2 induced profound cytoskeletal remodeling with formation of a thick ring of cortical actin within which clusters of CD36 could not aggregate, thereby attenuating binding of oxLDL to the surface of cells. By inhibiting recruitment of monocytes into early atherosclerotic lesions, and the subsequent binding and internalization of oxLDL by macrophages, Slit2 could represent a potent new tool to combat individual steps that collectively result in progression of atherosclerosis.


Assuntos
Aterosclerose/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lipídeos/imunologia , Lipoproteínas LDL/genética , Proteínas do Tecido Nervoso/genética , Animais , Aterosclerose/imunologia , Aterosclerose/patologia , Vasos Sanguíneos/imunologia , Antígenos CD36/genética , Antígenos CD36/imunologia , Modelos Animais de Doenças , Células Espumosas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipídeos/genética , Lipoproteínas LDL/imunologia , Macrófagos/imunologia , Camundongos , Monócitos/imunologia , Proteínas do Tecido Nervoso/metabolismo , Receptores Depuradores/genética , Receptores Depuradores/imunologia
9.
Nature ; 590(7844): 29-31, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33469204
10.
Immunity ; 54(3): 468-483.e5, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33484643

RESUMO

Tissue resident mast cells (MCs) rapidly initiate neutrophil infiltration upon inflammatory insult, yet the molecular mechanism is still unknown. Here, we demonstrated that MC-derived tumor necrosis factor (TNF) was crucial for neutrophil extravasation to sites of contact hypersensitivity-induced skin inflammation by promoting intraluminal crawling. MC-derived TNF directly primed circulating neutrophils via TNF receptor-1 (TNFR1) while being dispensable for endothelial cell activation. The MC-derived TNF was infused into the bloodstream by directional degranulation of perivascular MCs that were part of the vascular unit with access to the vessel lumen. Consistently, intravenous administration of MC granules boosted neutrophil extravasation. Pronounced and rapid intravascular MC degranulation was also observed upon IgE crosslinking or LPs challenge indicating a universal MC potential. Consequently, the directional MC degranulation of pro-inflammatory mediators into the bloodstream may represent an important target for therapeutic approaches aimed at dampening cytokine storm syndromes or shock symptoms, or intentionally pushing immune defense.


Assuntos
Vasos Sanguíneos/imunologia , Dermatite de Contato/imunologia , Inflamação/imunologia , Mastócitos/imunologia , Neutrófilos/imunologia , Pele/patologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Circulação Sanguínea , Degranulação Celular , Células Cultivadas , Doenças do Sistema Imunitário , Transtornos Leucocíticos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação de Neutrófilo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Vesículas Secretórias/metabolismo , Fator de Necrose Tumoral alfa/genética
11.
Front Immunol ; 12: 798211, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975909

RESUMO

The bones and joints in the skeletal system are composed of diverse cell types, including vascular niches, bone cells, connective tissue cells and mineral deposits and regulate whole-body homeostasis. The capacity of maintaining strength and generation of blood lineages lies within the skeletal system. Bone harbours blood and immune cells and their progenitors, and vascular cells provide several immune cell type niches. Blood vessels in bone are phenotypically and functionally diverse, with distinct capillary subtypes exhibiting striking changes with age. The bone vasculature has a special impact on osteogenesis and haematopoiesis, and dysregulation of the vasculature is associated with diverse blood and bone diseases. Ageing is associated with perturbed haematopoiesis, loss of osteogenesis, increased adipogenesis and diminished immune response and immune cell production. Endothelial and perivascular cells impact immune cell production and play a crucial role during inflammation. Here, we discuss normal and maladapted vascular niches in bone during development, homeostasis, ageing and bone diseases such as rheumatoid arthritis and osteoarthritis. Further, we discuss the role of vascular niches during bone malignancy.


Assuntos
Envelhecimento/imunologia , Vasos Sanguíneos/imunologia , Doenças Ósseas/imunologia , Osso e Ossos/irrigação sanguínea , Células-Tronco Hematopoéticas/imunologia , Articulações/irrigação sanguínea , Nicho de Células-Tronco , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Diferenciação Celular , Proliferação de Células , Células Progenitoras Endoteliais/imunologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Homeostase , Humanos , Osteoartrite/imunologia , Osteoartrite/metabolismo , Osteoartrite/patologia , Fenótipo
12.
Front Immunol ; 11: 2091, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072072

RESUMO

Macrophages play a central role in dictating the tissue response to infection and orchestrating subsequent repair of the damage. In this context, macrophages residing in the lungs continuously sense and discriminate among a wide range of insults to initiate the immune responses important to host-defense. Inflammatory tissue injury also leads to activation of proteases, and thereby the coagulation pathway, to optimize injury and repair post-infection. However, long-lasting inflammatory triggers from macrophages can impair the lung's ability to recover from severe injury, leading to increased lung vascular permeability and neutrophilic injury, hallmarks of Acute Lung Injury (ALI). In this review, we discuss the roles of toll-like receptor 4 (TLR4) and protease activating receptor 2 (PAR2) expressed on the macrophage cell-surface in regulating lung vascular inflammatory signaling.


Assuntos
Lesão Pulmonar Aguda/imunologia , Vasos Sanguíneos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Receptor PAR-2/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Vasos Sanguíneos/lesões , Vasos Sanguíneos/patologia , Permeabilidade Capilar/imunologia , Humanos , Pulmão/irrigação sanguínea , Macrófagos/patologia
13.
Exp Dermatol ; 29(11): 1046-1054, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32998178

RESUMO

Metastatic melanoma is a devastating disease with a marginal-albeit increasing-hope for cure. Melanoma has a high mutation rate which correlates to the expression of numerous neo-antigens and thus is associated with the potential to induce and strengthen effective antitumoral immunity. However, the incomplete and potentially insufficient response to established immunotherapies (response rates usually do not markedly exceed 60%) already points to the need of further studies to improve treatment strategies. Multiple tumor escape mechanisms that allow melanoma to evade from antitumoral immune responses have been characterized and must be overcome to achieve a better clinical efficacy of immunotherapies. Recently, promising progress has been made in targeting tumor vasculature to control and increase the infiltration of tumors with effector lymphocytes. It has been hypothesized that amplified lymphocytic infiltrates in melanoma metastases result in a switch of the tumor microenvironment from a non-inflammatory to an inflammatory state. In this view point essay, we discuss the requirements for successful homing of lymphocytes to melanoma tissue and we present a mouse melanoma xenograft model that allows the investigation of human tumor vessels in vivo. Furthermore, current clinical studies dealing with the activation of melanoma vasculature for enhanced effectiveness of immunotherapy protocols are presented and open questions for routine clinical application are addressed.


Assuntos
Endotélio/imunologia , Imunoterapia , Linfócitos/imunologia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Animais , Vasos Sanguíneos/imunologia , Movimento Celular , Modelos Animais de Doenças , Humanos , Melanoma/irrigação sanguínea , Melanoma/imunologia , Melanoma/secundário , Camundongos , Receptores de Retorno de Linfócitos , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Evasão Tumoral/imunologia
15.
Proc Natl Acad Sci U S A ; 117(39): 24316-24325, 2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32929010

RESUMO

Platelets are best known for their vasoprotective responses to injury and inflammation. Here, we have asked whether they also support vascular integrity when neither injury nor inflammation is present. Changes in vascular barrier function in dermal and meningeal vessels were measured in real time in mouse models using the differential extravasation of fluorescent tracers as a biomarker. Severe thrombocytopenia produced by two distinct methods caused increased extravasation of 40-kDa dextran from capillaries and postcapillary venules but had no effect on extravasation of 70-kDa dextran or albumin. This reduction in barrier function required more than 4 h to emerge after thrombocytopenia was established, reverting to normal as the platelet count recovered. Barrier dysfunction was also observed in mice that lacked platelet-dense granules, dense granule secretion machinery, glycoprotein (GP) VI, or the GPVI signaling effector phospholipase C (PLC) γ2. It did not occur in mice lacking α-granules, C type lectin receptor-2 (CLEC-2), or protease activated receptor 4 (PAR4). Notably, although both meningeal and dermal vessels were affected, intracerebral vessels, which are known for their tighter junctions between endothelial cells, were not. Collectively, these observations 1) highlight a role for platelets in maintaining vascular homeostasis in the absence of injury or inflammation, 2) provide a sensitive biomarker for detecting changes in platelet-dependent barrier function, 3) identify which platelet processes are required, and 4) suggest that the absence of competent platelets causes changes in the vessel wall itself, accounting for the time required for dysfunction to emerge.


Assuntos
Plaquetas/imunologia , Vasos Sanguíneos/imunologia , Hemostasia , Homeostase , Animais , Vasos Sanguíneos/lesões , Vasos Sanguíneos/fisiopatologia , Feminino , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Masculino , Meninges/irrigação sanguínea , Meninges/imunologia , Camundongos , Fosfolipase C gama/genética , Fosfolipase C gama/imunologia , Pele/irrigação sanguínea , Pele/imunologia
16.
J Thromb Thrombolysis ; 50(3): 567-579, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32627126

RESUMO

After the emergence of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and Middle East Respiratory Syndrome Coronavirus (MERS-CoV) in the last two decades, the world is facing its new challenge in Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic with unprecedented global response. With the expanding domain of presentations in COVID-19 patients, the full range of manifestations is yet to unfold. The classical clinical symptoms for SARS-CoV-2 affected patients are dry cough, high fever, dyspnoea, lethal pneumonia whereas many patients have also been found to be associated with a few additional signs and clinical manifestations of isolated vasculopathy. Albeit a deep and profound knowledge has been gained on the clinical features and management of COVID-19, less clear association has been provided on SARS-CoV-2 mediated direct or indirect vasculopathy and its possible correlation with disease prognosis. The accumulative evidences suggest that novel coronavirus, apart from its primary respiratory confinement, may also invade vascular endothelial cells of several systems including cerebral, cardio-pulmonary as well as renal microvasculature, modulating multiple visceral perfusion indices. Here we analyse the phylogenetic perspective of SARS-CoV-2 along with other strains of ß-coronaviridae from a standpoint of vasculopathic derangements. Based on the existing case reports, literature and open data bases, we also analyse the differential pattern of vasculopathy related changes in COVID-19 positive patients. Besides, we debate the need of modulation in clinical approach from a hemodynamical point of view, as a measure towards reducing disease transmission, morbidity and mortality in SARS-CoV-2 affected patients.


Assuntos
Betacoronavirus/patogenicidade , Vasos Sanguíneos/virologia , Infecções por Coronavirus/virologia , Pneumonia Viral/virologia , Doenças Vasculares/virologia , Animais , Betacoronavirus/genética , Betacoronavirus/imunologia , Coagulação Sanguínea , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatologia , COVID-19 , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/fisiopatologia , Citocinas/metabolismo , Hemodinâmica , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Pandemias , Filogenia , Pneumonia Viral/imunologia , Pneumonia Viral/metabolismo , Pneumonia Viral/fisiopatologia , Prognóstico , Medição de Risco , Fatores de Risco , SARS-CoV-2 , Doenças Vasculares/imunologia , Doenças Vasculares/metabolismo , Doenças Vasculares/fisiopatologia
17.
J Thromb Thrombolysis ; 50(3): 499-511, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32700024

RESUMO

The COVID-19 pandemic now totaling 13,000,000 cases and over 571,000 deaths has continued to teach the medical, scientific and lay communities about viral infectious disease in the modern era. Among the many lessons learned for the medical community is the potential for transmissibility and host infectivity of the SARS-CoV-2 virus. Moreover, it has become clear that the virus can affect any organ including the circulatory system, directly via either tissue tropism or indirectly stemming from inflammatory responses in the form of innate immunity, leukocyte debris such as cell-free DNA and histones and RNA viral particles. The following review considers COVID-19-associated vasculitis and vasculopathy as a defining feature of a virus-induced systemic disease with acute, subacute and potential chronic health implications.


Assuntos
Betacoronavirus/patogenicidade , Vasos Sanguíneos/virologia , Infecções por Coronavirus/virologia , Pneumonia Viral/virologia , Vasculite/virologia , Animais , Betacoronavirus/imunologia , Coagulação Sanguínea , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/sangue , Pandemias , Pneumonia Viral/diagnóstico , Pneumonia Viral/imunologia , Pneumonia Viral/fisiopatologia , Prognóstico , Fatores de Risco , SARS-CoV-2 , Vasculite/diagnóstico , Vasculite/imunologia , Vasculite/fisiopatologia
18.
Clin Immunol ; 217: 108493, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32526273

Assuntos
Complexo Antígeno-Anticorpo/biossíntese , Betacoronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Doenças do Complexo Imune/imunologia , Pneumonia Viral/imunologia , Síndrome Respiratória Aguda Grave/imunologia , Vasculite/imunologia , Anticorpos Antivirais/biossíntese , Complexo Antígeno-Anticorpo/efeitos dos fármacos , Betacoronavirus/imunologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Vasos Sanguíneos/virologia , COVID-19 , Complemento C3/antagonistas & inibidores , Complemento C3/biossíntese , Inativadores do Complemento/uso terapêutico , Infecções por Coronavirus/complicações , Infecções por Coronavirus/tratamento farmacológico , Infecções por Coronavirus/virologia , Síndrome da Liberação de Citocina/complicações , Síndrome da Liberação de Citocina/tratamento farmacológico , Síndrome da Liberação de Citocina/virologia , Humanos , Doenças do Complexo Imune/complicações , Doenças do Complexo Imune/tratamento farmacológico , Doenças do Complexo Imune/virologia , Imunidade Humoral/efeitos dos fármacos , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Interleucina-6/antagonistas & inibidores , Interleucina-6/biossíntese , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/tratamento farmacológico , Pneumonia Viral/virologia , Receptores de Interleucina-1/antagonistas & inibidores , Receptores de Interleucina-1/biossíntese , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/complicações , Síndrome Respiratória Aguda Grave/tratamento farmacológico , Síndrome Respiratória Aguda Grave/virologia , Índice de Gravidade de Doença , Vasculite/complicações , Vasculite/tratamento farmacológico , Vasculite/virologia
19.
Clin Immunol ; 217: 108487, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32479986

RESUMO

Coronavirus Disease 2019 (COVID-19) is an ongoing public health emergency and new knowledge about its immunopathogenic mechanisms is deemed necessary in the attempt to reduce the death burden, globally. For the first time in worldwide literature, we provide scientific evidence that in COVID-19 vasculitis a life-threatening escalation from type 2 T-helper immune response (humoral immunity) to type 3 hypersensitivity (immune complex disease) takes place. The subsequent deposition of immune complexes inside the vascular walls is supposed to induce a severe inflammatory state and a cytokine release syndrome, whose interleukin-6 is the key myokine, from the smooth muscle cells of blood vessels.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Doenças do Complexo Imune/imunologia , Pneumonia Viral/imunologia , Síndrome Respiratória Aguda Grave/imunologia , Células Th2/imunologia , Vasculite/imunologia , Idoso , Anticorpos Antivirais/biossíntese , Complexo Antígeno-Anticorpo/biossíntese , Betacoronavirus/imunologia , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Vasos Sanguíneos/virologia , COVID-19 , Complemento C3/biossíntese , Infecções por Coronavirus/complicações , Infecções por Coronavirus/virologia , Síndrome da Liberação de Citocina/complicações , Síndrome da Liberação de Citocina/virologia , Progressão da Doença , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Humanos , Doenças do Complexo Imune/complicações , Doenças do Complexo Imune/virologia , Imunidade Humoral , Imunoglobulina G/biossíntese , Imunoglobulina M/biossíntese , Interleucina-6/biossíntese , Masculino , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/complicações , Síndrome Respiratória Aguda Grave/virologia , Células Th2/patologia , Células Th2/virologia , Vasculite/complicações , Vasculite/virologia
20.
Yakugaku Zasshi ; 140(4): 509-512, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-32238633

RESUMO

The thymus is a vital organ for functional immune systems, and is the site of T cell development, which plays a central role in cellular immune defenses. Unlike other major organs, the thymus is highly dynamic in size and structure. It shrinks immediately upon bacterial infection, aging, pregnancy, mental stress, nutritional deficiency, and more. The reduction in size and function of the thymus during such biological events is called thymic involution or thymic atrophy; thymic involution is a particularly important issue because dysfunctional T cell immunity increases the risks of tumorigenesis and infectious diseases. However, the molecular mechanisms underlying thymic involution remain obscure. Our recent study indicated that blood vessels are remodeled during thymic involution that occurs upon aging, estradiol-treatment, or nutritional deficiency. We also found that prostanoid synthesis is induced during thymic involution. Treatment with non-steroidal anti-inflammatory drugs (NSAIDs), aspirin or etodolac, at least partially inhibited thymic involution-induced remodeling of the blood vessels, suggesting that prostanoids are involved in blood vessel remodeling. Our results revealed the potential role of blood vessel remodeling during thymic involution, which can lead to biological stress-induced immunosenescence.


Assuntos
Vasos Sanguíneos/imunologia , Vasos Sanguíneos/fisiopatologia , Estresse Fisiológico/imunologia , Estresse Psicológico/imunologia , Timo/imunologia , Remodelação Vascular , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Humanos , Camundongos , Prostaglandinas/metabolismo , Prostaglandinas/fisiologia , Estresse Fisiológico/fisiologia , Linfócitos T/imunologia , Timo/patologia , Remodelação Vascular/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA