Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.634
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 42(1): 427-53, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38360547

RESUMO

The role of the autoimmune regulator (Aire) in central immune tolerance and thymic self-representation was first described more than 20 years ago, but fascinating new insights into its biology continue to emerge, particularly in the era of advanced single-cell genomics. We briefly describe the role of human genetics in the discovery of Aire, as well as insights into its function gained from genotype-phenotype correlations and the spectrum of Aire-associated autoimmunity-including insights from patients with Aire mutations with broad and diverse implications for human health. We then highlight emerging trends in Aire biology, focusing on three topic areas. First, we discuss medullary thymic epithelial diversity and the role of Aire in thymic epithelial development. Second, we highlight recent developments regarding the molecular mechanisms of Aire and its binding partners. Finally, we describe the rapidly evolving biology of the identity and function of extrathymic Aire-expressing cells (eTACs), and a novel eTAC subset called Janus cells, as well as their potential roles in immune homeostasis.


Assuntos
Proteína AIRE , Autoimunidade , Fatores de Transcrição , Humanos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Animais , Timo/imunologia , Timo/metabolismo , Mutação , Tolerância Imunológica , Células Epiteliais/metabolismo , Células Epiteliais/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/metabolismo
2.
Cell ; 187(17): 4713-4732.e19, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-38968937

RESUMO

Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer.


Assuntos
Tolerância Imunológica , Progesterona , Progestinas , Inibidor 1 da Ativação de Células T com Domínio V-Set , Animais , Feminino , Inibidor 1 da Ativação de Células T com Domínio V-Set/metabolismo , Humanos , Camundongos , Gravidez , Progestinas/farmacologia , Progestinas/metabolismo , Progesterona/metabolismo , Neoplasias da Mama/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Receptores de Progesterona/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Placenta/imunologia
3.
Cell ; 175(2): 429-441.e16, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30245008

RESUMO

Targeting autophagy in cancer cells and in the tumor microenvironment are current goals of cancer therapy. However, components of canonical autophagy play roles in other biological processes, adding complexity to this goal. One such alternative function of autophagy proteins is LC3-associated phagocytosis (LAP), which functions in phagosome maturation and subsequent signaling events. Here, we show that impairment of LAP in the myeloid compartment, rather than canonical autophagy, induces control of tumor growth by tumor-associated macrophages (TAM) upon phagocytosis of dying tumor cells. Single-cell RNA sequencing (RNA-seq) analysis revealed that defects in LAP induce pro-inflammatory gene expression and trigger STING-mediated type I interferon responses in TAM. We found that the anti-tumor effects of LAP impairment require tumor-infiltrating T cells, dependent upon STING and the type I interferon response. Therefore, autophagy proteins in the myeloid cells of the tumor microenvironment contribute to immune suppression of T lymphocytes by effecting LAP.


Assuntos
Tolerância Imunológica/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Fagocitose/fisiologia , Animais , Autofagia/imunologia , Linhagem Celular , Interações Hospedeiro-Patógeno , Humanos , Tolerância Imunológica/imunologia , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Células Mieloides/metabolismo , Fagossomos/fisiologia , Linfócitos T/metabolismo , Microambiente Tumoral/fisiologia
4.
Immunity ; 54(5): 931-946.e11, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33838102

RESUMO

Activation of the STAT5 transcription factor downstream of the Interleukin-2 receptor (IL-2R) induces expression of Foxp3, a critical step in the differentiation of regulatory T (Treg) cells. Due to the pleiotropic effects of IL-2R signaling, it is unclear how STAT5 acts directly on the Foxp3 locus to promote its expression. Here, we report that IL-2 - STAT5 signaling converged on an enhancer (CNS0) during Foxp3 induction. CNS0 facilitated the IL-2 dependent CD25+Foxp3- precursor to Treg cell transition in the thymus. Its deficiency resulted in impaired Treg cell generation in neonates, which was partially mitigated with age. While the thymic Treg cell paucity caused by CNS0 deficiency did not result in autoimmunity on its own, it exacerbated autoimmune manifestations caused by disruption of the Aire gene. Thus, CNS0 enhancer activity ensures robust Treg cell differentiation early in postnatal life and cooperatively with other tolerance mechanisms minimizes autoimmunity.


Assuntos
Linhagem da Célula/imunologia , Fatores de Transcrição Forkhead/imunologia , Tolerância Imunológica/imunologia , Interleucina-2/imunologia , Linfócitos T Reguladores/imunologia , Animais , Autoimunidade/imunologia , Diferenciação Celular/imunologia , Elementos Facilitadores Genéticos/imunologia , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Camundongos , Receptores de Interleucina-2/imunologia , Fator de Transcrição STAT5/imunologia , Transdução de Sinais/imunologia
5.
Immunity ; 54(5): 947-961.e8, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33930308

RESUMO

The transcription factor Foxp3 plays crucial roles for Treg cell development and function. Conserved non-coding sequences (CNSs) at the Foxp3 locus control Foxp3 transcription, but how they developmentally contribute to Treg cell lineage specification remains obscure. Here, we show that among Foxp3 CNSs, the promoter-upstream CNS0 and the intergenic CNS3, which bind distinct transcription factors, were activated at early stages of thymocyte differentiation prior to Foxp3 promoter activation, with sequential genomic looping bridging these regions and the promoter. While deletion of either CNS0 or CNS3 partially compromised thymic Treg cell generation, deletion of both completely abrogated the generation and impaired the stability of Foxp3 expression in residual Treg cells. As a result, CNS0 and CNS3 double-deleted mice succumbed to lethal systemic autoimmunity and inflammation. Thus, hierarchical and coordinated activation of Foxp3 CNS0 and CNS3 initiates and stabilizes Foxp3 gene expression, thereby crucially controlling Treg cell development, maintenance, and consequently immunological self-tolerance.


Assuntos
Elementos Facilitadores Genéticos/imunologia , Fatores de Transcrição Forkhead/imunologia , Linfócitos T Reguladores/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas/imunologia , Tolerância a Antígenos Próprios/imunologia
6.
Immunol Rev ; 323(1): 303-315, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38501766

RESUMO

Besides its canonical role in protecting the host from pathogens, the immune system plays an arguably equally important role in maintaining tissue homeostasis. Within barrier tissues that interface with the external microenvironment, induction of immune tolerance to innocuous antigens, such as commensal, dietary, and environmental antigens, is key to establishing immune homeostasis. The early postnatal period represents a critical window of opportunity in which parallel development of the tissue, immune cells, and microbiota allows for reciprocal regulation that shapes the long-term immunological tone of the tissue and subsequent risk of immune-mediated diseases. During early infancy, the immune system appears to sacrifice pro-inflammatory functions, prioritizing the establishment of tissue tolerance. In this review, we discuss mechanisms underlying early life windows for intestinal tolerance with a focus on newly identified RORγt+ antigen-presenting cells-Thetis cells-and highlight the role of the intestinal microenvironment in shaping intestinal immune system development and tolerance.


Assuntos
Homeostase , Tolerância Imunológica , Mucosa Intestinal , Humanos , Animais , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Microbioma Gastrointestinal/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo
7.
Immunol Rev ; 326(1): 191-202, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39046826

RESUMO

Cow milk protein allergy (CMPA) is one of the most common food allergies in the pediatric age worldwide. Prevalence, persistence, and severity of this condition are on the rise, with a negative impact on the health-related quality of life of the patients and families and on the costs related to its management. Another relevant issue is that CMPA in early life may be the first stage of the "allergic march," leading to the occurrence of other atopic manifestations later in life, especially asthma, atopic eczema, urticaria, and rhinoconjunctivitis. Thus, "disease modification" options that are able to modulate the disease course of pediatric patients affected by CMPA would be very welcomed by affected families and healthcare systems. In this review, we report the most relevant progress on this topic.


Assuntos
Hipersensibilidade a Leite , Proteínas do Leite , Humanos , Hipersensibilidade a Leite/imunologia , Hipersensibilidade a Leite/terapia , Animais , Proteínas do Leite/imunologia , Bovinos , Alérgenos/imunologia , Criança , Qualidade de Vida
8.
Proc Natl Acad Sci U S A ; 121(41): e2404841121, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39348545

RESUMO

Severe and chronic infections, including pneumonia, sepsis, and tuberculosis (TB), induce long-lasting epigenetic changes that are associated with an increase in all-cause postinfectious morbidity and mortality. Oncology studies identified metabolic drivers of the epigenetic landscape, with the tricarboxylic acid (TCA) cycle acting as a central hub. It is unknown if the TCA cycle also regulates epigenetics, specifically DNA methylation, after infection-induced immune tolerance. The following studies demonstrate that lipopolysaccharide and Mycobacterium tuberculosis induce changes in DNA methylation that are mediated by the TCA cycle. Infection-induced DNA hypermethylation is mitigated by inhibitors of cellular metabolism (rapamycin, everolimus, metformin) and the TCA cycle (isocitrate dehydrogenase inhibitors). Conversely, exogenous supplementation with TCA metabolites (succinate and itaconate) induces DNA hypermethylation and immune tolerance. Finally, TB patients who received everolimus have less DNA hypermethylation demonstrating proof of concept that metabolic manipulation can mitigate epigenetic scars.


Assuntos
Ciclo do Ácido Cítrico , Metilação de DNA , Tolerância Imunológica , Lipopolissacarídeos , Mycobacterium tuberculosis , Tuberculose , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/imunologia , Humanos , Animais , Tuberculose/imunologia , Tuberculose/genética , Tuberculose/microbiologia , Camundongos , Epigênese Genética , Succinatos/metabolismo , Everolimo/farmacologia , Ácido Succínico/metabolismo
9.
Proc Natl Acad Sci U S A ; 121(31): e2404229121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39052836

RESUMO

The distinct human leukocyte antigen (HLA) class I expression pattern of human extravillous trophoblasts (EVT) endows them with unique tolerogenic properties that enable successful pregnancy. Nevertheless, how this process is elaborately regulated remains elusive. Previously, E74 like ETS transcription factor 3 (ELF3) was identified to govern high-level HLA-C expression in EVT. In the present study, ELF3 is found to bind to the enhancer region of two adjacent NOD-like receptor (NLR) genes, NLR family pyrin domain-containing 2 and 7 (NLRP2, NLRP7). Notably, our analysis of ELF3-deficient JEG-3 cells, a human choriocarcinoma cell line widely used to study EVT biology, suggests that ELF3 transactivates NLRP7 while suppressing the expression of NLRP2. Moreover, we find that NLRP2 and NLRP7 have opposing effects on HLA-C expression, thus implicating them in immune evasion at the maternal-fetal interface. We confirmed that NLRP2 suppresses HLA-C levels and described a unique role for NLRP7 in promoting HLA-C expression in JEG-3. These results suggest that these two NLR genes, which arose via gene duplication in primates, are fine-tuned by ELF3 yet have acquired divergent functions to enable proper expression levels of HLA-C in EVT, presumably through modulating the degradation kinetics of IkBα. Targeting the ELF3-NLRP2/NLRP7-HLA-C axis may hold therapeutic potential for managing pregnancy-related disorders, such as recurrent hydatidiform moles and fetal growth restriction, and thus improve placental development and pregnancy outcomes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Reguladoras de Apoptose , Trofoblastos Extravilosos , Antígenos HLA-C , Trofoblastos , Feminino , Humanos , Gravidez , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Antígenos HLA-C/metabolismo , Antígenos HLA-C/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Trofoblastos/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
10.
Proc Natl Acad Sci U S A ; 121(5): e2317762121, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38261616

RESUMO

Intravenous immunoglobulin (IVIg), a preparation of polyclonal serum IgG pooled from numerous blood donors, has been used for nearly three decades and is proving to be an efficient treatment for many autoimmune blistering diseases, including pemphigus vulgaris (PV). Despite its widespread use and therapeutic success, its mechanisms of action are not completely understood. Some of its anti-inflammatory and immunomodulatory actions have been studied. In this study, the authors present a twenty-year follow-up of 21 patients with clinical and immunopathological confirmed PV, treated with IVIg as monotherapy, according to an established published protocol. IVIg therapy produced long-term sustained, clinical, serological, and immunopathological remission. For 20 y, these patients received no drugs and experienced no disease. This observation suggests that there was the establishment of immune balance or restoration of immune regulation in these PV patients. Twelve (57%) patients experienced no relapse during follow-up. Six (29%) patients experienced a relapse due to acute stress or post-coronavirus infection and/or vaccination. Reinstitution of IVIg resulted in prompt sustained recovery. Three (14.2%) patients, in clinical and serological remission, died due to unrelated causes. No severe adverse effects from IVIg were documented in all 21 patients. The simultaneous or sequential anti-inflammatory and immunomodulatory effects of IVIg may have influenced the long-term clinical remission observed. This study provides a human prototype to examine the pathophysiology of autoimmunity and a model to study immune regulation and mechanisms that can facilitate restoring immune tolerance.


Assuntos
Doenças Autoimunes , Pênfigo , Humanos , Imunoglobulinas Intravenosas , Tolerância Imunológica , Anti-Inflamatórios
11.
Immunity ; 47(1): 107-117.e8, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28709804

RESUMO

Regulatory T (Treg) cells expressing the transcription factor Foxp3 are critical for the prevention of autoimmunity and the suppression of anti-tumor immunity. The major self-antigens recognized by Treg cells remain undefined, representing a substantial barrier to the understanding of immune regulation. Here, we have identified natural Treg cell ligands in mice. We found that two recurrent Treg cell clones, one prevalent in prostate tumors and the other associated with prostatic autoimmune lesions, recognized distinct non-overlapping MHC-class-II-restricted peptides derived from the same prostate-specific protein. Notably, this protein is frequently targeted by autoantibodies in experimental models of prostatic autoimmunity. On the basis of these findings, we propose a model in which Treg cell responses at peripheral sites converge on those self-proteins that are most susceptible to autoimmune attack, and we suggest that this link could be exploited as a generalizable strategy for identifying the Treg cell antigens relevant to human autoimmunity.


Assuntos
Autoantígenos/metabolismo , Epitopos de Linfócito T/metabolismo , Neoplasias da Próstata/imunologia , Linfócitos T Reguladores/imunologia , Timo/fisiologia , Animais , Autoanticorpos/metabolismo , Autoantígenos/genética , Autoantígenos/imunologia , Diferenciação Celular , Células Clonais , Mapeamento de Epitopos , Fatores de Transcrição Forkhead/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Ativação Linfocitária , Masculino , Camundongos
12.
Proc Natl Acad Sci U S A ; 120(7): e2208509120, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36745791

RESUMO

Antigenic peptides derived from introns are presented on major histocompatibility (MHC) class I molecules, but how these peptides are produced is poorly understood. Here, we show that an MHC class I epitope (SL8) sequence inserted in the second intron of the ß-globin gene in a C57BL/6 mouse (HBB) generates immune tolerance. Introduction of SL8-specific CD8+ T cells derived from OT-1 transgenic mice resulted in a threefold increase in OT-1 T cell proliferation in HBB animals, as compared to wild-type animals. The growth of MCA sarcoma cells expressing the intron-derived SL8 epitope was suppressed in wild-type animals compared to HBB mice. The ß-globin pre-mRNA was detected in the light polysomal fraction, and introducing stop codons identified a non-AUG initiation site between +228 and +255 nts upstream of the SL8. Isolation of ribosome footprints confirmed translation initiation within this 27 nt sequence. Furthermore, treatment with splicing inhibitor shifts the translation of the pre-mRNA to monosomal fractions and results in an increase of intron-derived peptide substrate as shown by polysome profiling and cell imaging. These results show that non-AUG-initiated translation of pre-mRNAs generates peptides for MHC class I immune tolerance and helps explain why alternative tissue-specific splicing is tolerated by the immune system.


Assuntos
Antígenos de Histocompatibilidade Classe I , Precursores de RNA , Animais , Camundongos , Antígenos de Histocompatibilidade Classe I/genética , Precursores de RNA/genética , Linfócitos T CD8-Positivos , Biossíntese de Proteínas , Apresentação de Antígeno , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Tolerância Imunológica/genética , Epitopos , Antígenos de Histocompatibilidade Classe II/genética
13.
Proc Natl Acad Sci U S A ; 120(25): e2218668120, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37307481

RESUMO

A longstanding goal has been to find an antigen-specific preventive therapy, i.e., a vaccine, for autoimmune diseases. It has been difficult to find safe ways to steer the targeting of natural regulatory antigen. Here, we show that the administration of exogenous mouse major histocompatibility complex class II protein bounding a unique galactosylated collagen type II (COL2) peptide (Aq-galCOL2) directly interacts with the antigen-specific TCR through a positively charged tag. This leads to expanding a VISTA-positive nonconventional regulatory T cells, resulting in a potent dominant suppressive effect and protection against arthritis in mice. The therapeutic effect is dominant and tissue specific as the suppression can be transferred with regulatory T cells, which downregulate various autoimmune arthritis models including antibody-induced arthritis. Thus, the tolerogenic approach described here may be a promising dominant antigen-specific therapy for rheumatoid arthritis, and in principle, for autoimmune diseases in general.


Assuntos
Artrite Reumatoide , Doenças Autoimunes , Animais , Camundongos , Vacinas de Subunidades Antigênicas , Linfócitos T Reguladores , Anticorpos
14.
Gastroenterology ; 167(1): 90-103, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38604542

RESUMO

The only proven treatment for celiac disease is adherence to a strict, lifelong, gluten-free diet. However, complete dietary gluten avoidance is challenging and a substantial number of patients do not respond fully, clinically, or histologically, despite their best efforts. As celiac disease is common and its central pathophysiology is well elucidated, it has become attractive for drug development to address the limitations of dietary treatment. Most efforts address nonresponsive celiac disease, defined as continued symptoms and/or signs of disease activity despite a gluten-free diet, and the more severe forms of refractory celiac disease, types I and II. An increasing spectrum of therapeutic approaches target defined mechanisms in celiac disease pathogenesis and some have advanced to current phase 2 and 3 clinical studies. We discuss these approaches in terms of potential efficiency, practicability, safety, and need, as defined by patients, regulatory authorities, health care providers, and payors.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Doença Celíaca/tratamento farmacológico , Humanos , Resultado do Tratamento , Fármacos Gastrointestinais/uso terapêutico , Fármacos Gastrointestinais/efeitos adversos , Fármacos Gastrointestinais/administração & dosagem , Animais
15.
Eur J Immunol ; 54(8): e2451046, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38778501

RESUMO

Chronic hepatitis B (CHB) virus infection, which can be divided into immune-tolerant (IT), immune-active (IA), inactive carrier (IC) phases, and HBeAg-negative hepatitis (ENEG), can induce liver cirrhosis and eventually hepatocellular carcinoma (HCC). CD3+CD56+ NKT-like cells play an important role in antiviral immune response. However, the mechanism of NKT-like cells to mediate immune tolerance remains largely elusive. In this study, we observed circulating NKT-like cells from IC and IT CHB patients were phenotypically and functionally impaired, manifested by increased expression of inhibitory receptor TIGIT and decreased capacity of secreting antiviral cytokines. Besides, TIGIT+ NKT-like cells of IC and IT CHB patients expressed lower levels of cytotoxic cytokines than the TIGIT- subset. Furthermore, increased expression of CD155, the ligand of TIGIT, on plasmacytoid dendritic cells (pDCs) was detected in IC and IT CHB patients. Importantly, the co-culture of NKT-like cells and pDCs showed that NKT-like cells restored their antiviral ability after TIGIT blockade upon HBV peptide stimulation in IC and IT CHB patients. In conclusion, our findings suggest that the TIGIT pathway may mediate immune tolerance in IT CHB patients and lead to functional impairment in IC patients, indicating that TIGIT may be a potential therapeutic checkpoint for immunotherapy of CHB patients.


Assuntos
Complexo CD3 , Antígeno CD56 , Células Dendríticas , Vírus da Hepatite B , Hepatite B Crônica , Tolerância Imunológica , Células T Matadoras Naturais , Receptores Imunológicos , Humanos , Receptores Imunológicos/metabolismo , Hepatite B Crônica/imunologia , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Antígeno CD56/metabolismo , Masculino , Vírus da Hepatite B/imunologia , Feminino , Células T Matadoras Naturais/imunologia , Adulto , Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Complexo CD3/imunologia , Complexo CD3/metabolismo , Pessoa de Meia-Idade , Receptores Virais/metabolismo , Receptores Virais/imunologia , Citocinas/metabolismo , Citocinas/imunologia
16.
Trends Immunol ; 43(2): 148-162, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35033428

RESUMO

Ubc13-catalyzed K63 ubiquitination is a major control point for immune signaling. Recent evidence has shown that the control of multiple immune functions, including chronic inflammation, pathogen responses, lymphocyte activation, and regulatory signaling, is altered by K63 ubiquitination. In this review, we detail the novel cellular sensors that are dependent on K63 ubiquitination for their function in the immune signaling network. Many pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can target K63 ubiquitination to inhibit pathogen immune responses; we describe novel details of the pathways involved and summarize recent clinically relevant SARS-CoV-2-specific responses. We also discuss recent evidence that regulatory T cell (Treg) versus T helper (TH) 1 and TH17 cell subset regulation might involve K63 ubiquitination. Knowledge gaps that merit future investigation and clinically relevant pathways are also addressed.


Assuntos
COVID-19 , Lisina , Humanos , Lisina/metabolismo , SARS-CoV-2 , Transdução de Sinais , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
17.
Immunity ; 44(2): 406-21, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26885861

RESUMO

Human CD4(+)CD25(hi)Foxp3(+)CD127(-) Treg and CD4(+)CD25(-)Foxp3(-) Tconv cell functions are governed by their metabolic requirements. Here we report a comprehensive comparative analysis between ex vivo human Treg and Tconv cells that comprises analyses of the proteomic networks in subcellular compartments. We identified a dominant proteomic signature at the metabolic level that primarily impacted the highly-tuned balance between glucose and fatty-acid oxidation in the two cell types. Ex vivo Treg cells were highly glycolytic while Tconv cells used predominantly fatty-acid oxidation (FAO). When cultured in vitro, Treg cells engaged both glycolysis and FAO to proliferate, while Tconv cell proliferation mainly relied on glucose metabolism. Our unbiased proteomic analysis provides a molecular picture of the impact of metabolism on ex vivo human Treg versus Tconv cell functions that might be relevant for therapeutic manipulations of these cells.


Assuntos
Ácidos Graxos/metabolismo , Glucose/metabolismo , Glicólise , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Antígenos CD4/metabolismo , Proliferação de Células , Células Cultivadas , Fatores de Transcrição Forkhead/metabolismo , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Oxirredução , Proteômica , Transcriptoma
18.
Rev Med Virol ; 34(4): e2570, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38964866

RESUMO

The question of whether patients in the immune-tolerant (IT) phase of chronic hepatitis B virus (HBV) infection should undergo antiviral therapy and determine the optimal regimen remains unclear. A comprehensive search of PubMed, Embase, MEDLINE, Cochrane Library, and Wanfang Data from inception to 5 December 2023, was conducted. Studies reporting on key outcomes such as HBV DNA undetectability, HBeAg loss or seroconversion, HBsAg loss or seroconversion, and hepatocellular carcinoma (HCC) incidence in patients in the IT phase of chronic HBV infection were included. In total, 23 studies were incorporated. Approximately 4% of patients in the IT phase achieved spontaneous HBeAg loss over 48 weeks of follow-up. Antiviral therapy demonstrated a favourable impact on HBV DNA negative conversion (Children: risk ratios [RR] = 6.83, 95% CI: 2.90-16.05; Adults: RR = 25.84, 95% CI: 6.47-103.31) and HBsAg loss rates (Children: RR = 9.49, 95% CI: 1.74-51.76; Adults: RR = 7.35, 95% CI: 1.41-38.27) for patients in the IT phase. Subgroup analysis revealed that in adult patients in the IT phase, interferon plus nucleos(t)ide analogues (NA)-treated patients exhibited a higher pooled rate of HBsAg loss or seroconversion than those treated with NA monotherapy (9% vs. 0%). Additionally, the pooled annual HCC incidence for patients in the IT phase was 3.03 cases per 1000 person-years (95% CI: 0.99-5.88). Adult patients in the IT phase had a significantly lower HCC incidence risk than HBeAg-positive indeterminate phase patients (RR = 0.46, 95% CI: 0.32-0.66), with no significant differences observed between IT and immune-active phases. Presently, there is insufficient evidence solely based on reducing the risk of HCC incidence, to recommend treating patients in the IT phase of chronic HBV infection. However, both adult and paediatric patients in the IT phase responded well to antiviral therapy, showing favourable rates of HBsAg loss or seroconversion.


Assuntos
Antivirais , Carcinoma Hepatocelular , Antígenos E da Hepatite B , Hepatite B Crônica , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/epidemiologia , Carcinoma Hepatocelular/virologia , Carcinoma Hepatocelular/imunologia , Hepatite B Crônica/complicações , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Hepatite B Crônica/epidemiologia , Hepatite B Crônica/imunologia , Neoplasias Hepáticas/epidemiologia , Neoplasias Hepáticas/virologia , Neoplasias Hepáticas/imunologia , Antivirais/uso terapêutico , Antígenos E da Hepatite B/sangue , Antígenos E da Hepatite B/imunologia , Vírus da Hepatite B/imunologia , Incidência , Antígenos de Superfície da Hepatite B/sangue , Antígenos de Superfície da Hepatite B/imunologia , DNA Viral/sangue , Tolerância Imunológica , Resultado do Tratamento , Soroconversão
19.
Brain ; 147(4): 1344-1361, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37931066

RESUMO

Neuromyelitis optica spectrum disorder (NMOSD) is a CNS autoimmune inflammatory disease mediated by T helper 17 (Th17) and antibody responses to the water channel protein, aquaporin 4 (AQP4), and associated with astrocytopathy, demyelination and axonal loss. Knowledge about disease pathogenesis is limited and the search for new therapies impeded by the absence of a reliable animal model. In our work, we determined that NMOSD is characterized by decreased IFN-γ receptor signalling and that IFN-γ depletion in AQP4201-220-immunized C57BL/6 mice results in severe clinical disease resembling human NMOSD. Pathologically, the disease causes autoimmune astrocytic and CNS injury secondary to cellular and humoral inflammation. Immunologically, the absence of IFN-γ allows for increased expression of IL-6 in B cells and activation of Th17 cells, and generation of a robust autoimmune inflammatory response. Consistent with NMOSD, the experimental disease is exacerbated by administration of IFN-ß, whereas repletion of IFN-γ, as well as therapeutic targeting of IL-17A, IL-6R and B cells, ameliorates it. We also demonstrate that immune tolerization with AQP4201-220-coupled poly(lactic-co-glycolic acid) nanoparticles could both prevent and effectively treat the disease. Our findings enhance the understanding of NMOSD pathogenesis and provide a platform for the development of immune tolerance-based therapies, avoiding the limitations of the current immunosuppressive therapies.


Assuntos
Neuromielite Óptica , Humanos , Animais , Camundongos , Neuromielite Óptica/patologia , Aquaporina 4 , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Linfócitos B , Autoanticorpos/metabolismo
20.
Mol Ther ; 32(10): 3453-3469, 2024 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-39192583

RESUMO

Antibody inhibitors pose an ongoing challenge to the treatment of subjects with inherited protein deficiency disorders, limiting the efficacy of both protein replacement therapy and corrective gene therapy. Beyond their central role as producers of serum antibody, B cells also exhibit many unique properties that could be exploited in cell therapy applications, notably including antigen-specific recognition and the linked capacity for antigen presentation. Here we employed CRISPR-Cas9 to demonstrate that ex vivo antigen-primed Blimp1-knockout "decoy" B cells, incapable of differentiation into plasma cells, participated in and downregulated host antigen-specific humoral responses after adoptive transfer. Following ex vivo antigen pulse, adoptively transferred high-affinity antigen-specific decoy B cells were diverted into germinal centers en masse, thereby reducing participation by endogenous antigen-specific B cells in T-dependent humoral responses and suppressing both cognate and linked antigen-specific immunoglobulin (Ig)G following immunization with conjugated antigen. This effect was dose-dependent and, importantly, did not impact concurrent unrelated antibody responses. We demonstrated the therapeutic potential of this approach by treating factor VIII (FVIII)-knockout mice with antigen-pulsed decoy B cells prior to immunization with an FVIII conjugate protein, thereby blunting the production of serum FVIII-specific IgG by an order of magnitude as well as reducing the proportion of animals exhibiting functional FVIII inhibition by 6-fold.


Assuntos
Formação de Anticorpos , Linfócitos B , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo , Animais , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Formação de Anticorpos/imunologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator 1 de Ligação ao Domínio I Regulador Positivo/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator VIII/imunologia , Fator VIII/genética , Sistemas CRISPR-Cas , Imunoglobulina G/imunologia , Transferência Adotiva , Humanos , Centro Germinativo/imunologia , Centro Germinativo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA