Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Innov Surg Sci ; 8(1): 3-8, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37842197

RESUMO

Objectives: Postoperative entero-enteric intussusception is a rare complication in adult patients with Crohn's disease (CD). The knowledge of this distinct complication and its timely diagnosis and therapy are of utmost importance to prevent fatal intestinal necrosis. There is no consensus about the optimal management of postoperative entero-enteric intussusception, although surgical exploration is widely advised. Case presentation: In this report we describe an unusual case of postoperative jejuno-jejunal intussusception following small bowel resection in a patient with stricturing CD. Furthermore, this report offers an overview of the available literature and summarizes the best approach and management strategies for adult intussusception associated with CD. Conclusions: Delay in diagnosis and therapy can lead to life-threatening complications. Early diagnosis and emergent surgical treatment prevent intestinal necrosis and reduce the risk of short bowel syndrome.

2.
Cell Calcium ; 116: 102802, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37757535

RESUMO

Chronic pancreatitis (CP) is a complex inflammatory disorder characterized by progressive fibrosis, leading to pancreatic dysfunction, reduced quality of life and an elevated pancreatic cancer risk. Current therapeutic options for CP are restricted to symptomatic treatment. Using ex vivo and in vivo preclinical disease models, Szabó et al. now explored for the first time the involvement of Store-operated Ca2+ entry (SOCE) in the progression of CP and propose that a selective pharmacological inhibition of the SOCE signaling component Orai1 might serve as specific treatment option for CP[1,2].


Assuntos
Cálcio , Pancreatite Crônica , Humanos , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Qualidade de Vida , Pancreatite Crônica/tratamento farmacológico , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/metabolismo
3.
Quant Imaging Med Surg ; 13(8): 4792-4805, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37581033

RESUMO

Background: Although there is growing evidence that functional involvement and structural changes of mesenteric adipose tissue (MAT) influence the course of Crohn's disease (CD), its viscoelastic properties remain elusive. Therefore, we aimed to investigate the viscoelastic properties of MAT in CD using magnetic resonance elastography (MRE), providing reference values for CD diagnosis. Methods: In this prospective proof-of-concept study, 31 subjects (CD: n=11; healthy controls: n=20) were consecutively enrolled in a specialized care center for inflammatory bowel diseases (tertiary/quaternary care). Inclusion criteria for the CD patients were a clinically and endoscopically established diagnosis of CD based on the clinical record, absence of other concurrent bowel diseases, scheduled surgery for the following day, and age of at least 18 years. Diagnoses were confirmed by histological analysis of the resected bowel the day after MRE. Subjects were investigated using MRE at 1.5-T with frequencies of 40-70 Hz. To retrieve shear wave speed (SWS), volumes of interest (VOIs) in MAT were drawn adjacent to CD lesions (MATCD) and on the opposite side without adjacent bowel lesions in patients (MATCD_Opp) and controls (MATCTRL). The presented study is not registered in the clinical trial platform. Results: A statistically significant decrease in mean SWS of 7% was found for MATCD_Opp vs. MATCTRL (0.76±0.05 vs. 0.82±0.04 m/s, P=0.012), whereas there was a nonsignificant trend with an 8% increase for MATCD vs. MATCD_Opp (0.82±0.07 vs. 0.76±0.05 m/s, P=0.098) and no difference for MATCD vs. MATCTRL. Preliminary area under the receiver operating characteristic curve (AUC) analysis showed diagnostic accuracy in detecting CD to be excellent for SWS of MATCD_Opp [AUC =0.82; 95% confidence interval (CI): 0.64-0.96] but poor for SWS of MATCD (AUC =0.52; 95% CI: 0.34-0.73). Conclusions: This study demonstrates the feasibility of MRE of MAT and presents preliminary reference values for CD patients and healthy controls. Our results motivate further studies for the biophysical characterization of MAT in inflammatory bowel disease.

4.
J Nat Prod ; 86(5): 1294-1306, 2023 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-37140218

RESUMO

Three new phenanthrene derivatives (1, 2, 4), one new fluorenone (3), and four known compounds (5-8) were isolated from the ethyl acetate extract of Dendrobium crumenatum Sw. stems using column chromatography. The chemical structures were elucidated by analysis of spectroscopic data. The absolute configuration of 4 was determined by electronic circular dichroism calculation. We also evaluated the immunomodulatory effects of compounds isolated from D. crumenatum in human peripheral blood mononuclear cells from healthy individuals and those from patients with multiple sclerosis in vitro. Dendrocrumenol B (2) and dendrocrumenol D (4) showed strong immunomodulatory effects on both CD3+ T cells and CD14+ monocytes. Compounds 2 and 4 could reduce IL-2 and TNF production in T cells and monocytes that were treated with phorbol-12-myristate-13-acetate and ionomycin (PMA/Iono). Deep immune profiling using high-dimensional single-cell mass cytometry could confirm immunomodulatory effects of 4, quantified by the reduction of activated T cell population under PMA/Iono stimulation, in comparison to the stimulated T cells without treatment.


Assuntos
Dendrobium , Fenantrenos , Humanos , Dendrobium/química , Leucócitos Mononucleares , Monócitos , Fenantrenos/farmacologia , Fenantrenos/química , Linfócitos T , Acetato de Tetradecanoilforbol/farmacologia , Fluorenos/química , Fluorenos/farmacologia
5.
Int J Mol Sci ; 24(6)2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36982601

RESUMO

Inflammatory bowel diseases (IBDs) consist of a group of chronic inflammatory disorders with a complex etiology, which represent a clinical challenge due to their often therapy-refractory nature. In IBD, inflammation of the intestinal mucosa is characterized by strong and sustained leukocyte infiltration, resulting in the loss of epithelial barrier function and subsequent tissue destruction. This is accompanied by the activation and the massive remodeling of mucosal micro-vessels. The role of the gut vasculature in the induction and perpetuation of mucosal inflammation is receiving increasing recognition. While the vascular barrier is considered to offer protection against bacterial translocation and sepsis after the breakdown of the epithelial barrier, endothelium activation and angiogenesis are thought to promote inflammation. The present review examines the respective pathological contributions of the different phenotypical changes observed in the microvascular endothelium during IBD, and provides an overview of potential vessel-specific targeted therapy options for the treatment of IBD.


Assuntos
Doenças Inflamatórias Intestinais , Mucosite , Humanos , Doenças Inflamatórias Intestinais/patologia , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Mucosite/patologia , Leucócitos/metabolismo
7.
EMBO J ; 42(6): e112202, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36795015

RESUMO

Lipids play a major role in inflammatory diseases by altering inflammatory cell functions, either through their function as energy substrates or as lipid mediators such as oxylipins. Autophagy, a lysosomal degradation pathway that limits inflammation, is known to impact on lipid availability, however, whether this controls inflammation remains unexplored. We found that upon intestinal inflammation visceral adipocytes upregulate autophagy and that adipocyte-specific loss of the autophagy gene Atg7 exacerbates inflammation. While autophagy decreased lipolytic release of free fatty acids, loss of the major lipolytic enzyme Pnpla2/Atgl in adipocytes did not alter intestinal inflammation, ruling out free fatty acids as anti-inflammatory energy substrates. Instead, Atg7-deficient adipose tissues exhibited an oxylipin imbalance, driven through an NRF2-mediated upregulation of Ephx1. This shift reduced secretion of IL-10 from adipose tissues, which was dependent on the cytochrome P450-EPHX pathway, and lowered circulating levels of IL-10 to exacerbate intestinal inflammation. These results suggest an underappreciated fat-gut crosstalk through an autophagy-dependent regulation of anti-inflammatory oxylipins via the cytochrome P450-EPHX pathway, indicating a protective effect of adipose tissues for distant inflammation.


Assuntos
Ácidos Graxos não Esterificados , Oxilipinas , Humanos , Adipócitos/metabolismo , Autofagia/fisiologia , Sistema Enzimático do Citocromo P-450/metabolismo , Sistema Enzimático do Citocromo P-450/farmacologia , Ácidos Graxos não Esterificados/metabolismo , Ácidos Graxos não Esterificados/farmacologia , Inflamação/genética , Inflamação/metabolismo , Interleucina-10/genética , Oxilipinas/metabolismo
8.
EMBO Mol Med ; 14(9): e15687, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35919953

RESUMO

Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune responses. Using mass cytometry (CyTOF) to analyze the immune cell composition in the lamina propria (LP) of patients with ulcerative colitis (UC) and Crohn's disease (CD), we observed an enrichment of CD4+ effector T cells producing IL-17A and TNF, CD8+ T cells producing IFNγ, T regulatory (Treg) cells, and innate lymphoid cells (ILC). The function of these immune cells is regulated by store-operated Ca2+ entry (SOCE), which results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI and STIM proteins. We observed that the pharmacologic inhibition of SOCE attenuated the production of proinflammatory cytokines including IL-2, IL-4, IL-6, IL-17A, TNF, and IFNγ by human colonic T cells and ILCs, reduced the production of IL-6 by B cells and the production of IFNγ by myeloid cells, but had no effect on the viability, differentiation, and function of intestinal epithelial cells. T cell-specific deletion of CRAC channel genes in mice showed that Orai1, Stim1, and Stim2-deficient T cells have quantitatively distinct defects in SOCE, which correlate with gradually more pronounced impairment of cytokine production by Th1 and Th17 cells and the severity of IBD. Moreover, the pharmacologic inhibition of SOCE with a selective CRAC channel inhibitor attenuated IBD severity and colitogenic T cell function in mice. Our data indicate that SOCE inhibition may be a suitable new approach for the treatment of IBD.


Assuntos
Canais de Cálcio Ativados pela Liberação de Cálcio , Doenças Inflamatórias Intestinais , Animais , Linfócitos T CD8-Positivos/metabolismo , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Humanos , Imunidade Inata , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Camundongos , Proteína ORAI1/metabolismo , Molécula 1 de Interação Estromal/genética , Células Th17/metabolismo
9.
BMC Infect Dis ; 22(1): 537, 2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35692034

RESUMO

BACKGROUND: Passive immunization against SARS-CoV-2 limits viral burden and death from COVID-19; however, it poses a theoretical risk of disease exacerbation through antibody-dependent enhancement (ADE). ADE after anti-SARS-CoV2 antibody treatment has not been reported, and therefore the potential risk and promoting factors remain unknown. CASE PRESENTATION: A 75-year-old female was admitted to the emergency room with recurrent, unexplained bruises and leukocytopenia, anemia, and thrombocytopenia. Evaluation of a bone marrow biopsy established the diagnosis of an acute promyelocytic leukemia (APL). SARS-CoV-2 RT-PCR testing of nasal and throat swabs on admission was negative. During the routine SARS-CoV-2 testing of inpatients, our patient tested positive for SARS-CoV-2 on day 14 after admission without typical COVID-19 symptoms. Due to disease- and therapy-related immunosuppression and advanced age conferring a high risk of progressing to severe COVID-19, casirivimab and imdevimab were administered as a preemptive approach. The patient developed immune activation and cytokine release syndrome (CRS) occurring within four hours of preemptive anti-SARS-CoV2 antibody (casirivimab/imdevimab) infusion. Immune activation and CRS were evidenced by a rapid increase in serum cytokines (IL-6, TNFα, IL-8, IL-10), acute respiratory insufficiency, and progressive acute respiratory distress syndrome. DISCUSSION AND CONCLUSION: The temporal relationship between therapeutic antibody administration and the rapid laboratory, radiological, and clinical deterioration suggests that CRS was an antibody-related adverse event, potentially exacerbated by APL treatment-mediated differentiation of leukemic blasts and promyelocytes. This case highlights the need for careful assessment of life-threatening adverse events after passive SARS-CoV-2 immunization, especially in the clinical context of patients with complex immune and hematological landscapes.


Assuntos
COVID-19 , Leucemia Promielocítica Aguda , Síndrome do Desconforto Respiratório , Idoso , Anticorpos Monoclonais Humanizados , COVID-19/complicações , COVID-19/diagnóstico , Teste para COVID-19 , Síndrome da Liberação de Citocina/diagnóstico , Síndrome da Liberação de Citocina/tratamento farmacológico , Síndrome da Liberação de Citocina/etiologia , Feminino , Humanos , Leucemia Promielocítica Aguda/complicações , Leucemia Promielocítica Aguda/diagnóstico , Leucemia Promielocítica Aguda/tratamento farmacológico , SARS-CoV-2
10.
J Clin Invest ; 132(9)2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35499080

RESUMO

Activated SUMOylation is a hallmark of cancer. Starting from a targeted screening for SUMO-regulated immune evasion mechanisms, we identified an evolutionarily conserved function of activated SUMOylation, which attenuated the immunogenicity of tumor cells. Activated SUMOylation allowed cancer cells to evade CD8+ T cell-mediated immunosurveillance by suppressing the MHC class I (MHC-I) antigen-processing and presentation machinery (APM). Loss of the MHC-I APM is a frequent cause of resistance to cancer immunotherapies, and the pharmacological inhibition of SUMOylation (SUMOi) resulted in reduced activity of the transcriptional repressor scaffold attachment factor B (SAFB) and induction of the MHC-I APM. Consequently, SUMOi enhanced the presentation of antigens and the susceptibility of tumor cells to CD8+ T cell-mediated killing. Importantly, SUMOi also triggered the activation of CD8+ T cells and thereby drove a feed-forward loop amplifying the specific antitumor immune response. In summary, we showed that activated SUMOylation allowed tumor cells to evade antitumor immunosurveillance, and we have expanded the understanding of SUMOi as a rational therapeutic strategy for enhancing the efficacy of cancer immunotherapies.


Assuntos
Apresentação de Antígeno , Neoplasias , Antígenos de Histocompatibilidade Classe I , Humanos , Evasão da Resposta Imune , Neoplasias/patologia , Sumoilação
13.
Mucosal Immunol ; 15(3): 480-490, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35169232

RESUMO

Immunosuppressive Interleukin (IL)-10 production by pro-inflammatory CD4+ T cells is a central self-regulatory function to limit aberrant inflammation. Still, the molecular mediators controlling IL-10 expression in human CD4+ T cells are largely undefined. Here, we identify a Notch/STAT3 signaling-module as a universal molecular switch to induce IL-10 expression across human naïve and major effector CD4+ T cell subsets. IL-10 induction was transient, jointly controlled by the transcription factors Blimp-1/c-Maf and accompanied by upregulation of several co-inhibitory receptors, including LAG-3, CD49b, PD-1, TIM-3 and TIGIT. Consistent with a protective role of IL-10 in inflammatory bowel diseases (IBD), effector CD4+ T cells from Crohn's disease patients were defective in Notch/STAT3-induced IL-10 production and skewed towards an inflammatory Th1/17 cell phenotype. Collectively, our data identify a Notch/STAT3-Blimp-1/c-Maf axis as a common anti-inflammatory pathway in human CD4+ T cells, which is defective in IBD and thus may represent an attractive therapeutic target.


Assuntos
Doença de Crohn , Doenças Inflamatórias Intestinais , Animais , Doença de Crohn/metabolismo , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-maf/genética , Proteínas Proto-Oncogênicas c-maf/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Th1/metabolismo
14.
Gastroenterology ; 162(3): 859-876, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34780721

RESUMO

BACKGROUND & AIMS: Monogenic forms of inflammatory bowel disease (IBD) illustrate the essential roles of individual genes in pathways and networks safeguarding immune tolerance and gut homeostasis. METHODS: To build a taxonomy model, we assessed 165 disorders. Genes were prioritized based on penetrance of IBD and disease phenotypes were integrated with multi-omics datasets. Monogenic IBD genes were classified by (1) overlapping syndromic features, (2) response to hematopoietic stem cell transplantation, (3) bulk RNA-sequencing of 32 tissues, (4) single-cell RNA-sequencing of >50 cell subsets from the intestine of healthy individuals and patients with IBD (pediatric and adult), and (5) proteomes of 43 immune subsets. The model was validated by addition of newly identified monogenic IBD defects. As a proof-of-concept, we explore the intersection between immunometabolism and antimicrobial activity for a group of disorders (G6PC3/SLC37A4). RESULTS: Our quantitative integrated taxonomy defines the cellular landscape of monogenic IBD gene expression across 102 genes with high and moderate penetrance (81 in the model set and 21 genes in the validation set). We illustrate distinct cellular networks, highlight expression profiles across understudied cell types (e.g., CD8+ T cells, neutrophils, epithelial subsets, and endothelial cells) and define genotype-phenotype associations (perianal disease and defective antimicrobial activity). We illustrate processes and pathways shared across cellular compartments and phenotypic groups and highlight cellular immunometabolism with mammalian target of rapamycin activation as one of the converging pathways. There is an overlap of genes and enriched cell-specific expression between monogenic and polygenic IBD. CONCLUSION: Our taxonomy integrates genetic, clinical and multi-omic data; providing a basis for genomic diagnostics and testable hypotheses for disease functions and treatment responses.


Assuntos
Doenças Inflamatórias Intestinais/classificação , Doenças Inflamatórias Intestinais/genética , Idade de Início , Antiporters/genética , Células Cultivadas , Classificação , Perfilação da Expressão Gênica , Estudos de Associação Genética , Genótipo , Glucose-6-Fosfatase/genética , Glucose-6-Fosfato/metabolismo , Humanos , Doenças Inflamatórias Intestinais/metabolismo , Macrófagos , Metabolômica , Proteínas de Transporte de Monossacarídeos/genética , Penetrância , Fenótipo , Transdução de Sinais/genética
16.
Mucosal Immunol ; 14(6): 1381-1392, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34420043

RESUMO

The SARS-CoV-2 pandemic has so far claimed over three and a half million lives worldwide. Though the SARS-CoV-2 mediated disease COVID-19 has first been characterized by an infection of the upper airways and the lung, recent evidence suggests a complex disease including gastrointestinal symptoms. Even if a direct viral tropism of intestinal cells has recently been demonstrated, it remains unclear, whether gastrointestinal symptoms are caused by direct infection of the gastrointestinal tract by SARS-CoV-2 or whether they are a consequence of a systemic immune activation and subsequent modulation of the mucosal immune system. To better understand the cause of intestinal symptoms we analyzed biopsies of the small intestine from SARS-CoV-2 infected individuals. Applying qRT-PCR and immunohistochemistry, we detected SARS-CoV-2 RNA and nucleocapsid protein in duodenal mucosa. In addition, applying imaging mass cytometry and immunohistochemistry, we identified histomorphological changes of the epithelium, which were characterized by an accumulation of activated intraepithelial CD8+ T cells as well as epithelial apoptosis and subsequent regenerative proliferation in the small intestine of COVID-19 patients. In summary, our findings indicate that intraepithelial CD8+ T cells are activated upon infection of intestinal epithelial cells with SARS-CoV-2, providing one possible explanation for gastrointestinal symptoms associated with COVID-19.


Assuntos
Linfócitos T CD8-Positivos/imunologia , COVID-19/imunologia , Duodeno/imunologia , Imunidade nas Mucosas , Enteropatias/imunologia , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/imunologia , Ativação Linfocitária , SARS-CoV-2/imunologia , Adulto , Idoso , Animais , Apoptose , Linfócitos T CD8-Positivos/virologia , COVID-19/patologia , COVID-19/virologia , Estudos de Casos e Controles , Proliferação de Células , Chlorocebus aethiops , Duodeno/patologia , Duodeno/virologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Enteropatias/patologia , Enteropatias/virologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Linfócitos Intraepiteliais/virologia , Masculino , Reepitelização , SARS-CoV-2/patogenicidade , Células Vero , Carga Viral
17.
Mucosal Immunol ; 14(3): 566-573, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33608656

RESUMO

Viral infections with SARS-CoV-2 can cause a multi-facetted disease, which is not only characterized by pneumonia and overwhelming systemic inflammatory immune responses, but which can also directly affect the digestive system and infect intestinal epithelial cells. Here, we review the current understanding of intestinal tropism of SARS-CoV-2 infection, its impact on mucosal function and immunology and summarize the effect of immune-suppression in patients with inflammatory bowel disease (IBD) on disease outcome of COVID-19 and discuss IBD-relevant implications for the clinical management of SARS-CoV-2 infected individuals.


Assuntos
COVID-19/complicações , COVID-19/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/imunologia , SARS-CoV-2/fisiologia , Biomarcadores , COVID-19/diagnóstico , COVID-19/virologia , Humanos , Imunidade Inata , Doenças Inflamatórias Intestinais/diagnóstico , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Índice de Gravidade de Doença , Avaliação de Sintomas , Tropismo Viral , Internalização do Vírus
18.
Inflamm Bowel Dis ; 26(1): 66-79, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31276162

RESUMO

Crohn's disease (CD) patients can be grouped into patients suffering from ileitis, ileocolitis, jejunoileitis, and colitis. The pathophysiological mechanism underlying this regional inflammation is still unknown. Although most murine models of inflammatory bowel disease (IBD) develop inflammation in the colon, there is an unmet need for novel models that recapitulate the spontaneous and fluctuating nature of inflammation as seen in CD. Recently, mice with an intestinal epithelial cell-specific deletion for Caspase-8 (Casp8ΔIEC mice), which are characterized by cell death-driven ileitis and disrupted Paneth cell homeostasis, have been identified as a novel model of CD-like ileitis. Here we uncovered that genetic susceptibility alone is sufficient to drive ileitis in Casp8ΔIEC mice. In sharp contrast, environmental factors, such as a disease-relevant microbial flora, determine colonic inflammation. Accordingly, depending on the microbial environment, isogenic Casp8ΔIEC mice either exclusively developed ileitis or suffered from pathologies in several parts of the gastrointestinal tract. Colitis in these mice was characterized by massive epithelial cell death, leading to spread of commensal gut microbes to the extra-intestinal space and hence an aberrant activation of the systemic immunity. We further uncovered that Casp8ΔIEC mice show qualitative and quantitative changes in the intestinal microbiome associated with an altered mucosal and systemic immune response. In summary, we identified that inflammation in this murine model of CD-like inflammation is characterized by an immune reaction, presumably directed against a disease-relevant microbiota in a genetically susceptible host, with impaired mucosal barrier function and bacterial clearance at the epithelial interface.


Assuntos
Doença de Crohn/microbiologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Ileíte/microbiologia , Mucosa Intestinal/microbiologia , Animais , Caspase 8 , Doença de Crohn/genética , Modelos Animais de Doenças , Predisposição Genética para Doença/genética , Ileíte/genética , Inflamação , Mucosa Intestinal/imunologia , Camundongos
19.
Front Immunol ; 10: 2718, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31849941

RESUMO

Epigenetic programs that control posttranslational modifications of histone proteins and DNA itself tightly regulate transcriptional networks determining the identity and function of CD8+ T cells. Chromatin-modifying enzymes such as histone acetyltransferases and deacetylases, represent key molecular determinants of the epigenetic imprinting of CD8+ T cells. The functions of these enzymes highly depend on the availability of key products of cellular metabolism pathways such as acetyl-CoA, NAD (Nicotinamide adenine dinucleotide) and SEM (S-adenosylmethionine), suggesting that there is a close crosstalk between the metabolic and the epigenetic regulation of CD8+ T cells. In this review, we will discuss the metabolic regulation of CD8+ T cell epigenetics during activation and differentiation. We will furthermore summarize how metabolic signals from the tumor microenvironment (TME) shape the epigenetic landscape of CD8+ T cells to better understand the mechanism underlying CD8+ T cell exhaustion in anti-tumor and anti-viral immunity, which might help to overcome limitations of current CD8+ T cell-based therapies.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Epigênese Genética/imunologia , Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Viroses/imunologia , Acetilcoenzima A/metabolismo , Animais , Linfócitos T CD8-Positivos/transplante , Diferenciação Celular , Senescência Celular , Epigenômica , Histonas/metabolismo , Humanos , Vigilância Imunológica , Ativação Linfocitária , Microambiente Tumoral
20.
Sci Rep ; 9(1): 19471, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31857644

RESUMO

Multiple sclerosis (MS) is an inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS). Studies in rodent models demonstrated an association of CNS-infiltrating monocyte-derived macrophages with disease severity. However, little is known about humans. Here, we performed an exploratory analysis of peripheral blood mononuclear cells (PBMCs) isolated from healthy controls and drug-naïve patients with early MS using multiplexed single-cell mass cytometry and algorithm-based data analysis. Two antibody panels comprising a total of 64 antibodies were designed to comprehensively analyse diverse immune cell populations, with particular emphasis on monocytes. PBMC composition and marker expression were overall similar between the groups. However, an increased abundance of CCR7+ and IL-6+ T cells was detected in early MS-PBMCs, whereas NFAT1hiT-bethiCD4+ T cells were decreased. Similarly, we detected changes in the subset composition of the CCR7+ and MIPßhi HLA-DR+ lymphocyte compartment. Only mild alterations were detected in monocytes/myeloid cells of patients with early MS, namely a decreased abundance of CD141hiIRF8hiCXCR3+CD68- dendritic cells. Unlike in Crohn's disease, no significant differences were found in the monocyte fraction of patients with early MS compared to healthy controls. This study provides a valuable resource for future studies designed to characterise and target diverse PBMC subsets in MS.


Assuntos
Leucócitos Mononucleares/imunologia , Esclerose Múltipla/imunologia , Adulto , Estudos de Casos e Controles , Feminino , Citometria de Fluxo/métodos , Voluntários Saudáveis , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Espectrometria de Massas/métodos , Pessoa de Meia-Idade , Esclerose Múltipla/sangue , Análise de Célula Única/métodos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA