Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 619
Filtrar
1.
Br J Pharmacol ; 181(12): 1695-1719, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38528718

RESUMO

Cardio-cerebrovascular diseases encompass pathological changes in the heart, brain and vascular system, which pose a great threat to health and well-being worldwide. Moreover, metabolic diseases contribute to and exacerbate the impact of vascular diseases. Inflammation is a complex process that protects against noxious stimuli but is also dysregulated in numerous so-called inflammatory diseases, one of which is atherosclerosis. Inflammation involves multiple organ systems and a complex cascade of molecular and cellular events. Numerous studies have shown that inflammation plays a vital role in cardio-cerebrovascular diseases and metabolic diseases. The absent in melanoma 2 (AIM2) inflammasome detects and is subsequently activated by double-stranded DNA in damaged cells and pathogens. With the assistance of the mature effector molecule caspase-1, the AIM2 inflammasome performs crucial biological functions that underpin its involvement in cardio-cerebrovascular diseases and related metabolic diseases: The production of interleukin-1 beta (IL-1ß), interleukin-18 (IL-18) and N-terminal pore-forming Gasdermin D fragment (GSDMD-N) mediates a series of inflammatory responses and programmed cell death (pyroptosis and PANoptosis). Currently, several agents have been reported to inhibit the activity of the AIM2 inflammasome and have the potential to be evaluated for use in clinical settings. In this review, we systemically elucidate the assembly, biological functions, regulation and mechanisms of the AIM2 inflammasome in cardio-cerebrovascular diseases and related metabolic diseases and outline the inhibitory agents of the AIM2 inflammasome as potential therapeutic drugs.


Assuntos
Proteínas de Ligação a DNA , Inflamassomos , Doenças Metabólicas , Humanos , Inflamassomos/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Doenças Metabólicas/tratamento farmacológico , Doenças Metabólicas/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/metabolismo , Doenças Vasculares/imunologia , Inflamação/metabolismo , Inflamação/tratamento farmacológico
2.
Pediatr Rheumatol Online J ; 21(1): 38, 2023 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-37087470

RESUMO

BACKGROUND: This study evaluated the efficacy and safety of baricitinib (Janus kinase-1/2 inhibitor), in adult and pediatric Japanese patients with Nakajo-Nishimura syndrome/chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (NNS/CANDLE), stimulator of interferon genes-associated vasculopathy with onset during infancy (SAVI), or Aicardi-Goutières syndrome (AGS). METHODS: A Phase 2/3, multicenter, open-label study (NCT04517253) was conducted across 52 weeks. Primary efficacy endpoint assessed the change in mean daily diary score (DDS) from baseline to the end of primary treatment period. Other efficacy endpoints included change in mean DDS to the end of maintenance period, daily corticosteroid use, Physician's Global Assessment of Disease Activity (PGA) scores, and daily symptom-specific score (DSSS) from baseline to primary and maintenance treatment periods. All treatment-emergent adverse events (TEAEs) that occurred postdosing were recorded. RESULTS: Overall, 9 patients (5 with NNS, 3 with SAVI, and 1 with AGS) were enrolled; 55.6% were females, mean age was 26 years, and mean corticosteroid use/weight was 0.2 mg/kg. At the end of primary treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.22) and SAVI (0.21) and increased in the patient with AGS (0.07). At the end of maintenance treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.18) and SAVI (0.27) and increased in the patient with AGS (0.04). Mean percent corticosteroid use decreased by 18.4% in 3 out of 5 patients with NNS/CANDLE and 62.9% in 1 out of 3 patients with SAVI. Mean PGA score decreased from baseline in patients with NNS/CANDLE (1.60), SAVI (1.33), and AGS (1.0), and mean DSSS improved from baseline. All patients reported ≥ 1 TEAE. Frequently reported AEs included BK polyomavirus detection (3; 33.3%), increased blood creatine phosphokinase (2; 22.2%), anemia (2; 22.2%), and upper respiratory tract infection (2; 22.2%). Three (33.3%) patients reported serious adverse events, 1 of which was related to study drug. One patient with SAVI died due to intracranial hemorrhage, which was not related to study drug. CONCLUSION: Baricitinib may offer a potential therapeutic option for patients with NNS/CANDLE, SAVI, and AGS, with a positive benefit/risk profile in a vulnerable patient population with multiple comorbidities. TRIAL REGISTRATION: NLM clinicaltrials.gov, NCT04517253 . Registered 18 August 2020.


Assuntos
População do Leste Asiático , Doenças Hereditárias Autoinflamatórias , Interferon Tipo I , Inibidores de Janus Quinases , Adulto , Criança , Feminino , Humanos , Masculino , Doenças Autoimunes do Sistema Nervoso/tratamento farmacológico , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/imunologia , População do Leste Asiático/genética , Dermatopatias/tratamento farmacológico , Dermatopatias/genética , Dermatopatias/imunologia , Resultado do Tratamento , Inibidores de Janus Quinases/uso terapêutico , Doenças Hereditárias Autoinflamatórias/tratamento farmacológico , Doenças Hereditárias Autoinflamatórias/genética , Doenças Hereditárias Autoinflamatórias/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Síndrome , Lipodistrofia/tratamento farmacológico , Lipodistrofia/genética , Lipodistrofia/imunologia , Febre , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/genética , Doenças Vasculares/imunologia , Glucocorticoides/efeitos adversos , Glucocorticoides/uso terapêutico
3.
Stem Cell Res ; 65: 102974, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36399927

RESUMO

We have successfully created induced pluripotent stem cells (iPSC) from patients carrying a heterozygous mutation in the gene encoding STING. The gain-of-function mutation leads to constitutive activation of STING which leads to the development of the disease STING-associated vasculopathy with onset in infancy (SAVI). The iPSC lines derived from the SAVI patitents are shown to be morphologically and phenotypically normal and have the potential to self renew and differentiate into the three germ layers. These iPSC provide a powerful tools to investigate the role of STING in the regulation of immune responses and vascular renegeration.


Assuntos
Imunidade , Células-Tronco Pluripotentes Induzidas , Doenças Vasculares , Humanos , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/patologia , Mutação com Ganho de Função , Doenças Vasculares/genética , Doenças Vasculares/imunologia
4.
Proc Natl Acad Sci U S A ; 119(25): e2202327119, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35696583

RESUMO

Pediatric patients with constitutively active mutations in the cytosolic double-stranded-DNA-sensing adaptor STING develop an autoinflammatory syndrome known as STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients have elevated interferon-stimulated gene expression and suffer from interstitial lung disease (ILD) with lymphocyte predominate bronchus-associated lymphoid tissue (BALT). Mice harboring SAVI mutations (STING V154M [VM]) that recapitulate human disease also develop lymphocyte-rich BALT. Ablation of either T or B lymphocytes prolongs the survival of SAVI mice, but lung immune aggregates persist, indicating that T cells and B cells can independently be recruited as BALT. VM T cells produced IFNγ, and IFNγR deficiency prolonged the survival of SAVI mice; however, T-cell-dependent recruitment of infiltrating myeloid cells to the lung was IFNγ independent. Lethally irradiated VM recipients fully reconstituted with wild type bone-marrow-derived cells still developed ILD, pointing to a critical role for VM-expressing radioresistant parenchymal and/or stromal cells in the recruitment and activation of pathogenic lymphocytes. We identified lung endothelial cells as radioresistant cells that express STING. Transcriptional analysis of VM endothelial cells revealed up-regulation of chemokines, proinflammatory cytokines, and genes associated with antigen presentation. Together, our data show that VM-expressing radioresistant cells play a key role in the initiation of lung disease in VM mice and provide insights for the treatment of SAVI patients, with implications for ILD associated with other connective tissue disorders.


Assuntos
Células Endoteliais , Doenças Pulmonares Intersticiais , Proteínas de Membrana , Linfócitos T , Doenças Vasculares , Animais , Criança , Células Endoteliais/imunologia , Células Endoteliais/efeitos da radiação , Mutação com Ganho de Função , Humanos , Interferon gama/genética , Interferon gama/metabolismo , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/imunologia , Depleção Linfocítica , Tecido Linfoide/imunologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Tolerância a Radiação , Linfócitos T/imunologia , Doenças Vasculares/genética , Doenças Vasculares/imunologia
5.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35163148

RESUMO

Increased soluble endoglin (sENG) has been observed in human brain arteriovenous malformations (bAVMs). In addition, the overexpression of sENG in concurrence with vascular endothelial growth factor (VEGF)-A has been shown to induce dysplastic vessel formation in mouse brains. However, the underlying mechanism of sENG-induced vascular malformations is not clear. The evidence suggests the role of sENG as a pro-inflammatory modulator, and increased microglial accumulation and inflammation have been observed in bAVMs. Therefore, we hypothesized that microglia mediate sENG-induced inflammation and endothelial cell (EC) dysfunction in bAVMs. In this study, we confirmed that the presence of sENG along with VEGF-A overexpression induced dysplastic vessel formation. Remarkably, we observed increased microglial activation around dysplastic vessels with the expression of NLRP3, an inflammasome marker. We found that sENG increased the gene expression of VEGF-A, pro-inflammatory cytokines/inflammasome mediators (TNF-α, IL-6, NLRP3, ASC, Caspase-1, and IL-1ß), and proteolytic enzyme (MMP-9) in BV2 microglia. The conditioned media from sENG-treated BV2 (BV2-sENG-CM) significantly increased levels of angiogenic factors (Notch-1 and TGFß) and pERK1/2 in ECs but it decreased the level of IL-17RD, an anti-angiogenic mediator. Finally, the BV2-sENG-CM significantly increased EC migration and tube formation. Together, our study demonstrates that sENG provokes microglia to express angiogenic/inflammatory molecules which may be involved in EC dysfunction. Our study corroborates the contribution of microglia to the pathology of sENG-associated vascular malformations.


Assuntos
Endoglina/administração & dosagem , Endotélio Vascular/patologia , Inflamação/patologia , Microglia/patologia , Neovascularização Patológica/patologia , Doenças Vasculares/patologia , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Animais , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/imunologia , Doenças Vasculares/metabolismo , Fator A de Crescimento do Endotélio Vascular/efeitos adversos
6.
Biochem Pharmacol ; 197: 114909, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35021044

RESUMO

Vascular endothelial cells are major participants in and regulators of immune responses and inflammation. Vascular endotheliitis is regarded as a host immune-inflammatory response of the endothelium forming the inner surface of blood vessels in association with a direct consequence of infectious pathogen invasion. Vascular endotheliitis and consequent endothelial dysfunction can be a principle determinant of microvascular failure, which would favor impaired perfusion, tissue hypoxia, and subsequent organ failure. Emerging evidence suggests the role of vascular endotheliitis in the pathogenesis of coronavirus disease 2019 (COVID-19) and its related complications. Thus, once initiated, vascular endotheliitis and resultant cytokine storm cause systemic hyperinflammation and a thrombotic phenomenon in COVID-19, leading to acute respiratory distress syndrome and widespread organ damage. Vascular endotheliitis also appears to be a contributory factor to vasculopathy and coagulopathy in sepsis that is defined as life-threatening organ dysfunction due to a dysregulated response of the host to infection. Therefore, protecting endothelial cells and reversing vascular endotheliitis may be a leading therapeutic goal for these diseases associated with vascular endotheliitis. In this review, we outline the etiological and pathogenic importance of vascular endotheliitis in infection-related inflammatory diseases, including COVID-19, and possible mechanisms leading to vascular endotheliitis. We also discuss pharmacological agents which may be now considered as potential endotheliitis-based treatment modalities for those diseases.


Assuntos
COVID-19/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , Doenças Vasculares/patologia , COVID-19/complicações , COVID-19/imunologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Glucocorticoides/farmacologia , Glucocorticoides/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/imunologia , Sepse/tratamento farmacológico , Sepse/etiologia , Sepse/imunologia , Sepse/patologia , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/etiologia , Doenças Vasculares/imunologia , Tratamento Farmacológico da COVID-19
7.
Cardiovasc Res ; 118(2): 372-385, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33483732

RESUMO

Nucleotide-binding oligomerization domain-like receptor family pyrin domain containing 3 (NLRP3) is an intracellular innate immune receptor that recognizes a diverse range of stimuli derived from pathogens, damaged or dead cells, and irritants. NLRP3 activation causes the assembly of a large multiprotein complex termed the NLRP3 inflammasome, and leads to the secretion of bioactive interleukin (IL)-1ß and IL-18 as well as the induction of inflammatory cell death termed pyroptosis. Accumulating evidence indicates that NLRP3 inflammasome plays a key role in the pathogenesis of sterile inflammatory diseases, including atherosclerosis and other vascular diseases. Indeed, the results of the Canakinumab Anti-inflammatory Thrombosis Outcome Study trial demonstrated that IL-1ß-mediated inflammation plays an important role in atherothrombotic events and suggested that NLRP3 inflammasome is a key driver of atherosclerosis. In this review, we will summarize the current state of knowledge regarding the role of NLRP3 inflammasome in vascular diseases, in particular in atherosclerosis, vascular injury, aortic aneurysm, and Kawasaki disease vasculitis, and discuss NLRP3 inflammasome as a therapeutic target for these disorders.


Assuntos
Artérias/metabolismo , Inflamassomos/metabolismo , Inflamação/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doenças Vasculares/metabolismo , Vasculite/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Artérias/efeitos dos fármacos , Artérias/imunologia , Artérias/patologia , Humanos , Inflamassomos/antagonistas & inibidores , Inflamassomos/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/patologia , Terapia de Alvo Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Transdução de Sinais , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/imunologia , Doenças Vasculares/patologia , Vasculite/tratamento farmacológico , Vasculite/imunologia , Vasculite/patologia
8.
J Atheroscler Thromb ; 29(3): 297-307, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34248111

RESUMO

Sterile chronic inflammation causes cardiometabolic disorders; however, the mechanisms are not fully understood. Previous studies have demonstrated the degradation of cells/tissues in the vasculature and metabolic organs in lifestyle-associated diseases, such as diabetes and hyperlipidemia, suggesting the release and/or accumulation of nucleic acids from damaged cells. DNA is indispensable for life; however, DNA fragments, especially those from pathogens, strongly induce inflammation by the activation of DNA sensors. Growing evidence suggests that DNA-sensing mechanisms, which are normally involved in self-defense against pathogens as the innate immune system, are associated with the progression of inflammatory diseases in response to endogenous DNA fragments. There are several types of DNA sensors in our bodies. Toll-like receptor 9 (TLR9)-one of the most studied DNA sensors-recognizes DNA fragments in endosome. In addition, stimulator of interferon genes (STING), which has recently been extensively investigated, recognizes cyclic GMP-AMP (cGAMP) generated from DNA fragments in the cytosol. Both TLR9 and STING are known to play pivotal roles in host defense as the innate immune system. However, recent studies have indicated that the activation of these DNA sensors in immune cells, such as macrophages, promotes inflammation leading to the development of vascular and metabolic diseases associated with lifestyle. In this review, we discuss recent advances in determining the roles of DNA sensors in these disease contexts. Revealing a novel mechanism of sterile chronic inflammation regulated by DNA sensors might facilitate clinical interventions for these health conditions.


Assuntos
DNA/imunologia , Imunidade Inata , Doenças Metabólicas/imunologia , Doenças Vasculares/imunologia , Animais , DNA/metabolismo , Humanos , Doenças Metabólicas/metabolismo , Transdução de Sinais , Doenças Vasculares/metabolismo
9.
J Leukoc Biol ; 111(2): 301-312, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34730257

RESUMO

Deficiency of adenosine deaminase 2 (DADA2) is a monogenic vasculitis syndrome caused by autosomal-recessive loss-of-function mutations in the ADA2 gene (previously known as CECR1). Vasculitis, vasculopathy, and inflammation are dominant clinical features of this disease; the spectrum of manifestations includes immunodeficiency and lymphoproliferation as well as hematologic manifestations. ADA2 is primarily secreted by stimulated monocytes and macrophages. Aberrant monocyte differentiation to macrophages and neutrophils are important in the pathogenesis of DADA2, but little is known about T lymphocytes in this disease. We performed combined single-cell RNA sequencing and single-cell TCR sequencing in order to profile T cell repertoires in 10 patients with DADA2. Although there were no significant alterations of T cell subsets, we observed activation of both CD8+ and CD4+ T cells. There was no clonal expansion of T cells: most TCRs were expressed at basal levels in patients and healthy donors. TCR usage was private to individual patients and not disease specific, indicating as unlikely a common pathogenic background or predisposition to a common pathogen. We recognized activation of IFN pathways as a signature of T cells and STAT1 as a hub gene in the gene network of T cell activation and cytotoxicity. Overall, T cells in DADA2 patients showed distinct cell-cell interactions with monocytes, as compared with healthy donors, and many of these ligand-receptor interactions likely drove up-regulation of STAT1 in both T cells and other immune cells in patients. Our analysis reveals previously undercharacterized cell characteristics in DADA2.


Assuntos
Adenosina Desaminase/deficiência , Biomarcadores/metabolismo , Regulação da Expressão Gênica , Síndromes de Imunodeficiência/patologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Dermatopatias/patologia , Linfócitos T/patologia , Doenças Vasculares/patologia , Adenosina Desaminase/genética , Adolescente , Adulto , Estudos de Casos e Controles , Células Cultivadas , Criança , Feminino , Seguimentos , Perfilação da Expressão Gênica , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Mutação , Prognóstico , Fator de Transcrição STAT1/genética , Análise de Célula Única , Dermatopatias/genética , Dermatopatias/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/imunologia , Adulto Jovem
10.
Int Immunopharmacol ; 101(Pt B): 108334, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34768128

RESUMO

BACKGROUND: Cigarette smoke (CS) is associated with vascular injury and dysfunction, which may be mediated by iNOS and NLRP3. However, the exact mechanism is unknown. METHODS: iNOS-knockout and NLRP3-knockout C57BL/6 mice were exposed to air or CS. The vascular structure was examined by hematoxylin-eosin staining. The vascular tension was measured by a vascular reactivity assay. The expression of iNOS, NLRP3, caspase-1p20, IL-1ß and eNOS were measured by western blotting. Human aortic endothelial cells (HAECs) were exposed to L-NIL (iNOS inhibitor), MCC950 (NLRP3 inhibitor), ODQ (sGC inhibitor), KT5823 (PKG inhibitor) or TAPI-1 (TACE/ADAM17 inhibitor) for 1 h prior to cigarette smoke extract (CSE) treatment. The cell viability and lactate dehydrogenase activity were assessed and pyroptosis was determined by scanning electron microscopy. The mRNA expression of TNF-α, and protein expression of iNOS, active-TACE, NLRP3, caspase-1p20, IL-1ß, and eNOS were measured. RESULTS: CS resulted in shrinkage of endothelial cells, impaired aorta relaxation, reduced eNOS expression, and induced expression of iNOS, NLRP3, caspase-1p20 and IL-1ß, which could be prevented by knockdown of iNOS and NLRP3. CSE reduced cell viability, induced LDH release and pyroptosis, and promoted iNOS, NLRP3, caspase-1p20, and IL-1ß expression and reduced eNOS reduction, which could be reversed by inhibition of iNOS or NLRP3 in HAECs. Altogether, activation of the NLRP3 inflammasome by iNOS in CS-exposed HAECs may be mediated by the sGC/cGMP/PKG/TACE/TNF- α pathway. CONCLUSION: These results link iNOS to NLRP3 in CSE-stimulated HAECs through the sGC/cGMP/PKG/TACE/TNF-α pathway. The findings identify a mechanism through which iNOS and NLRP3 contribute to the pathogenesis of CS-induced pyroptosis and impaired aorta relaxation in HAECs.


Assuntos
Aorta/patologia , Fumar Cigarros/efeitos adversos , Células Endoteliais/fisiologia , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Doenças Vasculares/imunologia , Proteína ADAM17/metabolismo , Animais , Linhagem Celular , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Piroptose , Transdução de Sinais , Guanilil Ciclase Solúvel/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Int J Immunopathol Pharmacol ; 35: 20587384211042115, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34541915

RESUMO

BACKGROUND: Hypercoagulability is a risk factor of thromboembolic events in COVID-19. Anti-phospholipid (aPL) antibodies have been hypothesized to be involved. Typical COVID-19 dermatological manifestations of livedo reticularis and digital ischemia may resemble cutaneous manifestations of anti-phospholipid syndrome (APS). OBJECTIVES: To investigate the association between aPL antibodies and thromboembolic events, COVID-19 severity, mortality, and cutaneous manifestations in patients with COVID-19. METHODS: aPL antibodies [anti-beta2-glycoprotein-1 (B2GP1) and anti-cardiolipin (aCL) antibodies] were titered in frozen serum samples from hospitalized COVID-19 patients and the patients' clinical records were retrospectively analyzed. RESULTS: 173 patients were enrolled. aPL antibodies were detected in 34.7% of patients, anti-B2GP1 antibodies in 30.1%, and aCL antibodies in 10.4%. Double positivity was observed in 5.2% of patients. Thromboembolic events occurred in 9.8% of patients, including 11 pulmonary embolisms, 1 case of celiac tripod thrombosis, and six arterial ischemic events affecting the cerebral, celiac, splenic, or femoral-popliteal arteries or the aorta. aPL antibodies were found in 52.9% of patients with vascular events, but thromboembolic events were not correlated to aPL antibodies (adjusted OR = 1.69, p = 0.502). Ten patients (5.8%) had cutaneous signs of vasculopathy: nine livedo reticularis and one acrocyanosis. No significant association was observed between the presence of cutaneous vasculopathy and aPL antibodies (p = 0.692). CONCLUSIONS: Anti-phospholipid antibodies cannot be considered responsible for hypercoagulability and thrombotic events in COVID-19 patients. In COVID-19 patients, livedo reticularis and acrocyanosis do not appear to be cutaneous manifestations of APS.


Assuntos
Anticorpos Antifosfolipídeos/sangue , COVID-19/complicações , SARS-CoV-2 , Dermatopatias/sangue , Doenças Vasculares/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Anticardiolipina/sangue , COVID-19/sangue , COVID-19/imunologia , COVID-19/mortalidade , Feminino , Hospitalização , Humanos , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Estudos Soroepidemiológicos , Dermatopatias/imunologia , Dermatopatias/mortalidade , Doenças Vasculares/imunologia , Doenças Vasculares/mortalidade , beta 2-Glicoproteína I/imunologia
12.
Int Arch Allergy Immunol ; 182(11): 1103-1112, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34583360

RESUMO

BACKGROUND: It is plausible that gene polymorphisms in tumor necrosis factor-α (TNF-α), interleukin (IL)-1, IL-6, IL-8, and IL-18 may affect predisposition to microvascular complications of diabetes mellitus (DM), but the results of the so far published studies remain controversial. OBJECTIVES: We conducted this meta-analysis to clarify relationships between TNF-α/IL-1/IL-4/IL-8/IL-18 polymorphisms and predisposition to microvascular complications of DM by pooling the findings of eligible studies. METHODS: A comprehensive search of PubMed, Embase, Web of Science, and CNKI was endorsed by us to identify already published studies. Forty-nine studies were found to be eligible for the meta-analyses. RESULTS: The pooled meta-analyses results showed that genotypic frequencies of TNF-α -238 G/A, TNF-α -308 G/A, TNF-α -1,031 T/C, IL-1A -889 C/T, IL-1B -511 C/T, IL-6 -572 G/C, and IL-18 -137 G/C polymorphisms among patients with diabetic nephropathy (DN) and controls differed significantly. Moreover, genotypic frequencies of TNF-α -238 G/A and IL-8 -251 A/T polymorphisms among patients with diabetic retinopathy (DR) and controls also differed significantly. CONCLUSIONS: This meta-analysis suggested that TNF-α -238 G/A, TNF-α -308 G/A, TNF-α -1,031 T/C, IL-1A -889 C/T, IL-1B -511 C/T, IL-6 -572 G/C, and IL-18 -137 G/C polymorphisms may affect predisposition of DN. Moreover, TNF-α -238 G/A and IL-8 -251 A/T polymorphisms may affect predisposition of DR.


Assuntos
Citocinas/genética , Complicações do Diabetes/genética , Doenças Vasculares/genética , Complicações do Diabetes/imunologia , Predisposição Genética para Doença , Humanos , Microvasos , Doenças Vasculares/etiologia , Doenças Vasculares/imunologia
13.
Front Immunol ; 12: 559925, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33776989

RESUMO

Sickle Cell Anemia (SCA) is the most common genetic disorder around the world. The mutation in the ß-globin gene is responsible for a higher hemolysis rate, with further involvement of immunological molecules, especially cytokines, chemokines, growth factors, and anaphylatoxins. These molecules are responsible for inducing and attracting immune cells into circulation, thus contributing to increases in leukocytes and other pro-inflammatory mediators, and can culminate in a vaso-occlusive crisis (VOC). This study aimed to characterize the levels of these molecules in SCA patients in different clinical conditions in order to identify potential hallmarks of inflammation in these patients. An analytical prospective study was conducted using the serum of SCA patients in steady-state (StSt; n = 27) and VOC (n = 22), along with 53 healthy donors (HD). Samples from the VOC group were obtained on admission and on discharge, in the convalescent phase (CV). Levels of chemokines (CXCL8, CXCL10, CL2, CLL3, CCL4, CL5, and CCL11), cytokines (IL-1ß, IL-1ra, IL-2, IL-4, IL-5, IL-6, IL-7, IL-10, IL-12p70, IL-13, IL-17A, TNF-α, and IFN-γ) and growth factors (VEGF, FGFb, PDGF-BB, GM-CSF, and G-CSF) were measured using a Luminex assay, and anaphylatoxins (C3a, C4a, and C5a) were measured using Cytometric Bead Array. SCA patients in StSt showed a pro-inflammatory profile, and were indicated as being higher producers of CCL2, IL-1ß, IL-12p70, IFN-γ, IL-17A, and GM-CSF, while VOC is highlighted by molecules IL-4 and IL-5, but also IL-2, IL-7, PDGF-BB, and G-CSF. PDGF-BB and IL-1ra seemed to be two important hallmarks for the acute-to-chronic stage, due to their significant decrease after crisis inflammation and statistical difference in VOC and CV groups. These molecules show higher levels and a strong correlation with other molecules in VOC. Furthermore, they remain at higher levels even after crisis recovery, which suggest their importance in the role of inflammation during crisis and participation in immune cell adhesion and activation. These results support a relevant role of cytokines, neutrophil and monocytes, since these may act as markers of VOC inflammation in SCA patients.


Assuntos
Anemia Falciforme/imunologia , Citocinas/imunologia , Mediadores da Inflamação/imunologia , Inflamação/imunologia , Doenças Vasculares/imunologia , Adolescente , Adulto , Anemia Falciforme/metabolismo , Quimiocinas/imunologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Masculino , Modelos Imunológicos , Monócitos/imunologia , Monócitos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Mapas de Interação de Proteínas/imunologia , Doenças Vasculares/metabolismo , Adulto Jovem
14.
Rheumatology (Oxford) ; 60(12): 5509-5516, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33744916

RESUMO

In patients with SSc, the coexistence of ANCA-associated vasculitis (SSc-AAV) has been reported to be associated with a severe disease course, including significant pulmonary and renal involvement. The presence of ANCA is not uncommon in patients with SSc, and therefore clinicians must maintain a high index of clinical suspicion about SSc-AAV. p-ANCA and anti-myeloperoxidase antibodies are the most common antibodies observed. Patients typically present with clinical features of microscopic polyangiitis or renal-limited vasculitis. There are multiple areas of potential interaction in the pathogenesis of SSc and AAV, which can exacerbate/compound vascular disease. In addition, similar patterns of major internal organ involvement (e.g. lung and kidneys) are seen in both conditions. We highlight a diagnostic approach to SSc-AAV and the paucity of data to inform management. As such, SSc-AAV is typically treated as per isolated AAV, which can potentially be hazardous in patients with SSc (e.g. due to the association between high-dose steroid and scleroderma renal crisis). We propose that this rare clinical entity warrants rigorous investigation, including definition of a therapeutic strategy to ameliorate the potentially devastating combination of pathologies in SSc-AAV.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/complicações , Anticorpos Anticitoplasma de Neutrófilos/imunologia , Escleroderma Sistêmico/complicações , Doenças Vasculares/complicações , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/diagnóstico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/imunologia , Humanos , Gravidade do Paciente , Escleroderma Sistêmico/diagnóstico , Escleroderma Sistêmico/imunologia , Doenças Vasculares/diagnóstico , Doenças Vasculares/imunologia
15.
Nephrol Dial Transplant ; 36(4): 581-586, 2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-31755918

RESUMO

Immunoglobulin A nephropathy (IgAN) is considered as mesangiopathy since it initiates in the mesangium; however, other glomerular components are involved and the glomerular capillary wall offers the first contact to circulating macromolecular IgA1. Acute and active forms of IgAN are associated with endocapillary hypercellularity and vascular damage of various degrees, in severe cases with microangiopathy (MA) without or with thrombosis [thrombotic microangiopathy (TMA)]. Vascular damage activates complement and coagulation cascades. A defective complement regulation has recently been detected in active and progressive cases of IgAN. C4d deposits in renal biopsies have been found to be an early risk factor. These observations have raised interest in manifestation of MA and TMA in progressive cases of IgAN. MA-TMA lesions have been found in various percentages (2-53%) of patients with IgAN according to patients' selection and pathology definition of TMA. The association with hypertension (HTN) was so strong that it led to the hypothesis that MA/TMA in IgAN was a mere consequence of severe HTN. Old and new clinical and experimental data indicate that in IgAN the interaction of the glomerular capillary wall with immune reactants and complement uncontrolled activation leading to C4b deposits favours the development of MA-TMA, which plays a role in progression and renal function decline. The central role of complement activation is relevant also for the new therapeutic interventions offered by the pharma.


Assuntos
Ativação do Complemento/imunologia , Complemento C4b/imunologia , Glomerulonefrite por IGA/patologia , Glomérulos Renais/patologia , Microangiopatias Trombóticas/patologia , Doenças Vasculares/patologia , Glomerulonefrite por IGA/imunologia , Humanos , Glomérulos Renais/imunologia , Microangiopatias Trombóticas/imunologia , Doenças Vasculares/imunologia
16.
Clin Exp Dermatol ; 46(3): 525-531, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32986882

RESUMO

BACKGROUND: Direct immunofluorescence (DIF) findings in patients with livedoid vasculopathy (LV) may have benefits for disease differentiation when clinical presentations and/or histopathological findings are inconclusive. AIM: To investigate DIF findings in patients with a clinical and histopathological diagnosis of LV. METHODS: DIF findings of 62 patients with LV were analysed, and the published literature in the PubMed database was also reviewed and summarized. RESULTS: This study demonstrated deposition of immunoreactants in blood vessels (BVs) in 59 of the 62 patients (95.2%), and almost all cases were positive for a combination of multiple immunoreactants. Complement C3 and IgM formed the most common combination. The most common pattern was deposition in BVs and at the dermoepidermal junction (DEJ) (59.3%), followed by deposition in BVs alone (40.7%). Immunoreactant deposition in BVs involved superficial BVs with or without deep BVs. The median age of patients with positive DIF findings was significantly higher than that of patients with negative DIF findings for LV (P < 0.03). More recent lesions (present for < 6 months) had a significantly higher percentage of positive results than older lesions (present for ≥ 6 months) (85.2% vs. 14.8%, respectively; P < 0.001). CONCLUSION: In both the present study and in the published literature, DIF study in patients with LV showed positive immunoreactants ranging from 42.9% to 100%. C3 and IgM were the most common immunoreactants deposited in BVs, while the most common pattern was immunoreactant deposition in BVs and at the DEJ. Older patients and those with more recent lesions (< 6 months) had a significantly higher percentage of positive DIF results for LV than did younger patients and those with older lesions (≥ 6 months).


Assuntos
Doenças Vasculares/imunologia , Adulto , Complemento C3/análise , Feminino , Técnica Direta de Fluorescência para Anticorpo , Humanos , Imunoglobulina A/análise , Imunoglobulina G/análise , Imunoglobulina M/análise , Masculino , Pessoa de Meia-Idade , Pele/irrigação sanguínea
17.
CEN Case Rep ; 10(1): 59-63, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32776181

RESUMO

We report the first case of intravascular large B-cell lymphoma (IVLBCL) presenting with vasculitis-like symptoms and elevated serum levels of anti-neutrophil cytoplasmic antibody (ANCA) diagnosed by renal biopsy. The patient exhibited low-grade fever, systemic inflammatory reactions, multiple lung lesions, and persistent proteinuria, which were closely correlated with changes in serum titers of proteinase-3- and myeloperoxidase-ANCA. Preemptive therapy with prednisolone alone partially attenuated these symptoms. Renal biopsy did not reveal histopathological findings suggestive of granulomatous or microscopic polyangiitis. Glomerular and peritubular capillaries were diffusely occluded by CD20-positive large atypical mononuclear cells, with focal foot process effacement of podocytes in the glomeruli. Based on the specific immunophenotype of infiltrated atypical cells, the patient was diagnosed with IVLBCL. Chemotherapy regimens for IVLBCL improved clinical symptoms and led to remission of proteinuria. The ANCA titers decreased in parallel with reductions in the serum levels of the soluble interleukin-2 receptor, suggestive of an association between changes in ANCA levels and IVLBCL-related vascular injuries.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/sangue , Rim/patologia , Linfoma Difuso de Grandes Células B/diagnóstico , Doenças Vasculares/patologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biópsia , Ciclofosfamida/administração & dosagem , Ciclofosfamida/uso terapêutico , Doxorrubicina/administração & dosagem , Doxorrubicina/uso terapêutico , Feminino , Humanos , Injeções Espinhais , Rim/imunologia , Rim/ultraestrutura , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Linfoma Difuso de Grandes Células B/imunologia , Pessoa de Meia-Idade , Peroxidase/sangue , Prednisona/administração & dosagem , Prednisona/uso terapêutico , Proteinúria/diagnóstico , Proteinúria/etiologia , Rituximab/administração & dosagem , Rituximab/uso terapêutico , Resultado do Tratamento , Doenças Vasculares/imunologia , Vincristina/administração & dosagem , Vincristina/uso terapêutico
18.
Front Immunol ; 11: 532177, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193304

RESUMO

Background: Microparticles (MPs) are vesicular structures that derive from multiple cellular sources. MPs play important roles in intercellular communication, regulation of cell signaling or initiation of enzymatic processes. While MPs were characterized in Systemic Sclerosis (SSc) patients, their contribution to SSc pathogenesis remains unknown. Our aim was to investigate the potential role of MPs in SSc pathophysiology and their impact on tissue fibrosis. Methods: Ninety-six SSc patients and 37 sex-matched healthy donors (HD) were enrolled in this study in order to quantify and phenotype their plasmatic MPs by flow cytometry. The ability of MPs purified from SSc patients and HD controls to modulate fibroblast's extra-cellular matrix genes expression was evaluated in vitro by reverse transcriptase quantitative polymerase chain reaction. Results: SSc patients exhibited a higher concentration of circulatory MPs compared to HD. This difference was exacerbated when we only considered patients that were not treated with methotrexate or targeted disease-modifying antirheumatic drugs. Total circulatory MPs were associated to interstitial lung disease, lung fibrosis and diminished lung functional capacity, but also to vascular involvement such as active digital ulcers. Finally, contrary to HD MPs, MPs from SSc patients stimulated the production of extracellular matrix by fibroblast, demonstrating their profibrotic potential. Conclusions: In this study, we provide evidence for a direct profibrotic role of MPs from SSc patients, underpinned by strong clinical associations in a large cohort of patients.


Assuntos
Micropartículas Derivadas de Células/imunologia , Fibrose Pulmonar/imunologia , Escleroderma Sistêmico/imunologia , Doenças Vasculares/imunologia , Idoso , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/patologia , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/imunologia , Feminino , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Citometria de Fluxo , Regulação da Expressão Gênica/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Fibrose Pulmonar/sangue , Fibrose Pulmonar/patologia , Escleroderma Sistêmico/sangue , Escleroderma Sistêmico/patologia , Doenças Vasculares/sangue , Doenças Vasculares/patologia
19.
Front Immunol ; 11: 575219, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33133092

RESUMO

STING-associated vasculopathy with onset in infancy (SAVI) is an autosomal dominant disorder due to gain-of-function mutations in STING1, also known as TMEM173, encoding for STING. It was reported as a vasculopathy of infancy. However, since its description a wider spectrum of associated manifestations and disease-onset has been observed. We report a kindred with a heterozygous STING mutation (p.V155M) in which the 19-year-old proband suffered from isolated adult-onset ANCA-associated vasculitis. His father suffered from childhood-onset pulmonary fibrosis and renal failure attributed to ANCA-associated vasculitis, and died at the age of 30 years due to respiratory failure. In addition, an overview of the phenotypic spectrum of SAVI is provided highlighting (a) a high phenotypic variability with in some cases isolated manifestations, (b) the potential of adult-onset disease, and (c) a novel manifestation with ANCA-associated vasculitis.


Assuntos
Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/genética , Proteínas de Membrana/genética , Mutação , Doenças Vasculares/genética , Idade de Início , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/diagnóstico , Vasculite Associada a Anticorpo Anticitoplasma de Neutrófilos/imunologia , Predisposição Genética para Doença , Hereditariedade , Heterozigoto , Humanos , Masculino , Linhagem , Fenótipo , Prognóstico , Doenças Vasculares/diagnóstico , Doenças Vasculares/imunologia , Adulto Jovem
20.
Clin Exp Immunol ; 202(2): 193-209, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32978971

RESUMO

Innate immune sensing of viral molecular patterns is essential for development of antiviral responses. Like many viruses, SARS-CoV-2 has evolved strategies to circumvent innate immune detection, including low cytosine-phosphate-guanosine (CpG) levels in the genome, glycosylation to shield essential elements including the receptor-binding domain, RNA shielding and generation of viral proteins that actively impede anti-viral interferon responses. Together these strategies allow widespread infection and increased viral load. Despite the efforts of immune subversion, SARS-CoV-2 infection activates innate immune pathways inducing a robust type I/III interferon response, production of proinflammatory cytokines and recruitment of neutrophils and myeloid cells. This may induce hyperinflammation or, alternatively, effectively recruit adaptive immune responses that help clear the infection and prevent reinfection. The dysregulation of the renin-angiotensin system due to down-regulation of angiotensin-converting enzyme 2, the receptor for SARS-CoV-2, together with the activation of type I/III interferon response, and inflammasome response converge to promote free radical production and oxidative stress. This exacerbates tissue damage in the respiratory system, but also leads to widespread activation of coagulation pathways leading to thrombosis. Here, we review the current knowledge of the role of the innate immune response following SARS-CoV-2 infection, much of which is based on the knowledge from SARS-CoV and other coronaviruses. Understanding how the virus subverts the initial immune response and how an aberrant innate immune response contributes to the respiratory and vascular damage in COVID-19 may help to explain factors that contribute to the variety of clinical manifestations and outcome of SARS-CoV-2 infection.


Assuntos
Betacoronavirus/fisiologia , Infecções por Coronavirus/imunologia , Hipóxia/imunologia , Pneumonia Viral/imunologia , Doenças Vasculares/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Coagulação Sanguínea , COVID-19 , Humanos , Evasão da Resposta Imune , Imunidade Inata , Interferon Tipo I/metabolismo , Pandemias , Peptidil Dipeptidase A/metabolismo , Sistema Renina-Angiotensina , SARS-CoV-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...