Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.213
Filtrar
1.
JCI Insight ; 9(9)2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38716734

RESUMO

mRNA vaccines are likely to become widely used for the prevention of infectious diseases in the future. Nevertheless, a notable gap exists in mechanistic data, particularly concerning the potential effects of sequential mRNA immunization or preexisting immunity on the early innate immune response triggered by vaccination. In this study, healthy adults, with or without documented prior SARS-CoV-2 infection, were vaccinated with the BNT162b2/Comirnaty mRNA vaccine. Prior infection conferred significantly stronger induction of proinflammatory and type I IFN-related gene signatures, serum cytokines, and monocyte expansion after the prime vaccination. The response to the second vaccination further increased the magnitude of the early innate response in both study groups. The third vaccination did not further increase vaccine-induced inflammation. In vitro stimulation of PBMCs with TLR ligands showed no difference in cytokine responses between groups, or before or after prime vaccination, indicating absence of a trained immunity effect. We observed that levels of preexisting antigen-specific CD4 T cells, antibody, and memory B cells correlated with elements of the early innate response to the first vaccination. Our data thereby indicate that preexisting memory formed by infection may augment the innate immune activation induced by mRNA vaccines.


Assuntos
Vacina BNT162 , Vacinas contra COVID-19 , COVID-19 , Citocinas , Imunidade Inata , SARS-CoV-2 , Vacinação , Humanos , Imunidade Inata/imunologia , COVID-19/imunologia , COVID-19/prevenção & controle , SARS-CoV-2/imunologia , Adulto , Masculino , Vacina BNT162/imunologia , Vacina BNT162/administração & dosagem , Feminino , Vacinas contra COVID-19/imunologia , Vacinas contra COVID-19/administração & dosagem , Vacinação/métodos , Citocinas/imunologia , Vacinas de mRNA/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Pessoa de Meia-Idade , Linfócitos T CD4-Positivos/imunologia , Adulto Jovem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem
2.
Adv Protein Chem Struct Biol ; 140: 381-417, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38762276

RESUMO

Cardiovascular disease (CVD) and cancer are major contributors to global morbidity and mortality. This book chapter delves into the intricate relationship between the immune system and the pathogenesis of both cardiovascular and cancer diseases, exploring the roles of innate and adaptive immunities, immune regulation, and immunotherapy in these complex conditions. The innate immune system acts as the first line of defense against tissue damage and infection, with a significant impact on the initiation and progression of CVD and cancer. Endothelial dysfunction, a hallmark in CVD, shares commonalities with the tumor microenvironment in cancer, emphasizing the parallel involvement of the immune system in both conditions. The adaptive immune system, particularly T cells, contributes to prolonged inflammation in both CVD and cancer. Regulatory T cells and the intricate balance between different T cell subtypes influence disease progression, wound healing, and the outcomes of ischemic injury and cancer immunosurveillance. Dysregulation of immune homeostasis can lead to chronic inflammation, contributing to the development and progression of both CVD and cancer. Thus, immunotherapy emerged as a promising avenue for preventing and managing these diseases, with strategies targeting immune cell modulation, cytokine manipulation, immune checkpoint blockade, and tolerance induction. The impact of gut microbiota on CVD and cancer too is explored in this chapter, highlighting the role of gut leakiness, microbial metabolites, and the potential for microbiome-based interventions in cardiovascular and cancer immunotherapies. In conclusion, immunomodulatory strategies and immunotherapy hold promise in reshaping the landscape of cardiovascular and cancer health. Additionally, harnessing the gut microbiota for immune modulation presents a novel approach to prevent and manage these complex diseases, emphasizing the importance of personalized and precision medicine in healthcare. Ongoing research and clinical trials are expected to further elucidate the complex immunological underpinnings of CVD and cancer thereby refining these innovative approaches.


Assuntos
Doenças Cardiovasculares , Neoplasias , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Doenças Cardiovasculares/imunologia , Imunoterapia , Imunidade Inata/imunologia , Microbioma Gastrointestinal/imunologia , Animais , Imunidade Adaptativa/imunologia
3.
Immunity ; 57(5): 1160-1176.e7, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38697118

RESUMO

Multimodal single-cell profiling methods can capture immune cell variations unfolding over time at the molecular, cellular, and population levels. Transforming these data into biological insights remains challenging. Here, we introduce a framework to integrate variations at the human population and single-cell levels in vaccination responses. Comparing responses following AS03-adjuvanted versus unadjuvanted influenza vaccines with CITE-seq revealed AS03-specific early (day 1) response phenotypes, including a B cell signature of elevated germinal center competition. A correlated network of cell-type-specific transcriptional states defined the baseline immune status associated with high antibody responders to the unadjuvanted vaccine. Certain innate subsets in the network appeared "naturally adjuvanted," with transcriptional states resembling those induced uniquely by AS03-adjuvanted vaccination. Consistently, CD14+ monocytes from high responders at baseline had elevated phospho-signaling responses to lipopolysaccharide stimulation. Our findings link baseline immune setpoints to early vaccine responses, with positive implications for adjuvant development and immune response engineering.


Assuntos
Linfócitos B , Vacinas contra Influenza , Análise de Célula Única , Humanos , Vacinas contra Influenza/imunologia , Linfócitos B/imunologia , Centro Germinativo/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vacinação , Anticorpos Antivirais/imunologia , Adjuvantes Imunológicos , Adjuvantes de Vacinas , Monócitos/imunologia , Polissorbatos , Esqualeno/imunologia , Imunidade Inata/imunologia
4.
Immunity ; 57(5): 927-929, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38749392

RESUMO

Humans do not respond equally to vaccination. To investigate why, Mulè et al. developed a multimodal framework and found that high responders after unadjuvanted influenza vaccination exist in a naturally adjuvanted state, mimicking innate immunophenotypes following AS03-adjuvanted vaccination. This highlights biological factors that set apart high-antibody responders and how adjuvants can boost innate immune cues to improve humoral immunity.


Assuntos
Imunidade Inata , Vacinas contra Influenza , Humanos , Vacinas contra Influenza/imunologia , Imunidade Inata/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Vacinação , Adjuvantes Imunológicos , Imunidade Humoral , Adjuvantes de Vacinas , Anticorpos Antivirais/imunologia , Animais
5.
Immunity ; 57(5): 957-972, 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38749398

RESUMO

Platelets prevent blood loss during vascular injury and contribute to thrombus formation in cardiovascular disease. Beyond these classical roles, platelets are critical for the host immune response. They guard the vasculature against pathogens via specialized receptors, intracellular signaling cascades, and effector functions. Platelets also skew inflammatory responses by instructing innate immune cells, support adaptive immunosurveillance, and influence antibody production and T cell polarization. Concomitantly, platelets contribute to tissue reconstitution and maintain vascular function after inflammatory challenges. However, dysregulated activation of these multitalented cells exacerbates immunopathology with ensuing microvascular clotting, excessive inflammation, and elevated risk of macrovascular thrombosis. This dichotomy underscores the critical importance of precisely defining and potentially modulating platelet function in immunity.


Assuntos
Plaquetas , Imunidade Inata , Plaquetas/imunologia , Humanos , Animais , Imunidade Inata/imunologia , Inflamação/imunologia , Imunidade Adaptativa/imunologia , Trombose/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia
6.
Mol Immunol ; 170: 26-34, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38603988

RESUMO

Neutrophils represent an important asset of innate immunity. Neutrophils express myeloperoxidase (MPO) which is a heme-containing peroxidase involved in microbial killing. In this study, by using real-time quantitative PCR and Western blot analysis, the flounder MPO (PoMPO) was observed to be highly expressed in the head kidney, followed by spleen, gill, and intestine during ontogeny - during developmental stages from larvae to adults. Furthermore, PoMPO positive cells were present in major immune organs of flounder at all developmental stages, and the number of neutrophils was generally higher as the fish grew to a juvenile stage. In addition, flow cytometry analysis revealed that the proportion of PoMPO positive cells relative to leukocytes, in the peritoneal cavity, head kidney, and peripheral blood of flounder juvenile stage was 18.3 %, 34.8 %, and 6.0 %, respectively, which is similar to the adult stage in flounder as previously reported. The presence and tissue distribution of PoMPO during ontogeny suggests that PoMPO positive cells are indeed a player of the innate immunity at all developmental stages of flounder.


Assuntos
Linguado , Imunidade Inata , Neutrófilos , Peroxidase , Animais , Linguado/imunologia , Peroxidase/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Imunidade Inata/imunologia , Brânquias/imunologia , Rim Cefálico/imunologia , Proteínas de Peixes/metabolismo , Proteínas de Peixes/imunologia , Proteínas de Peixes/genética , Citometria de Fluxo , Baço/imunologia
7.
Mol Immunol ; 170: 131-143, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663254

RESUMO

Mammalian reovirus (MRV) is a non-enveloped, gene segmented double-stranded RNA (dsRNA) virus. It is an important zoonotic pathogen that infects many mammals and vertebrates that act as natural hosts and causes respiratory and digestive tract diseases. Studies have reported that RIG-I and MDA5 in the innate immune cytoplasmic RNA-sensing RIG-like receptor (RLR) signaling pathway can recognize dsRNA from MRV and promote antiviral type I interferon (IFN) responses. However, the mechanism by which many MRV-encoded proteins evade the host innate immune response remains unclear. Here, we show that exogenous µ1 protein promoted the proliferation of MRV in vitro, while knockdown of MRV µ1 protein expression by shRNA could impair MRV proliferation. Specifically, µ1 protein inhibited MRV or poly(I:C)-induced IFN-ß expression, and attenuated RIG-I/MDA5-mediated signaling axis transduction during MRV infection. Importantly, we found that µ1 protein significantly decreased IFN-ß mRNA expression induced by MDA5, RIG-I, MAVS, TBK1, IRF3(5D), and degraded the protein expression of exogenous MDA5, RIG-I, MAVS, TBK1 and IRF3 via the proteasomal and lysosomal pathways. Additionally, we show that µ1 protein can physically interact with MDA5, RIG-I, MAVS, TBK1, and IRF3 and attenuate the RIG-I/MDA5-mediated signaling cascades by blocking the phosphorylation and nuclear translocation of IRF3. In conclusion, our findings reveal that MRV outer capsid protein µ1 is a key factor in antagonizing RLRs signaling cascades and provide new strategies for effective prevention and treatment of MRV infection.


Assuntos
Proteína DEAD-box 58 , Fator Regulador 3 de Interferon , Helicase IFIH1 Induzida por Interferon , Orthoreovirus de Mamíferos , Receptores Imunológicos , Transdução de Sinais , Helicase IFIH1 Induzida por Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Proteína DEAD-box 58/metabolismo , Transdução de Sinais/imunologia , Humanos , Fosforilação , Orthoreovirus de Mamíferos/imunologia , Orthoreovirus de Mamíferos/fisiologia , Células HEK293 , Interferon beta/metabolismo , Interferon beta/imunologia , Animais , Núcleo Celular/metabolismo , Infecções por Reoviridae/imunologia , Proteínas Virais/metabolismo , Transporte Ativo do Núcleo Celular , Imunidade Inata/imunologia , Proteínas Serina-Treonina Quinases
8.
Cytokine ; 179: 156623, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38685155

RESUMO

Systemic lupus erythematosus (SLE) is a connective tissue disorder that affects various body systems. Both the innate and adaptive immunity contribute to the onset and progression of SLE. The main mechanism of SLE is an excessive immune response of immune cells to autoantigens, which leads to systemic inflammation and inflammation-induced organ damage. Notably, a subset of innate immune cells known as innate lymphoid cells (ILCs) has recently emerged. ILCs are pivotal in the early stages of infection; participate in immune responses, inflammation, and tissue repair; and regulate the immune function of the body by resisting pathogens and regulating autoimmune inflammation and metabolic homeostasis. Thus, ILCs dysfunction can lead to autoimmune diseases. This review discusses the maturation of ILCs, the potential mechanisms by which ILCs exacerbate SLE pathogenesis, and their contributions to organ inflammatory deterioration in SLE.


Assuntos
Imunidade Inata , Inflamação , Lúpus Eritematoso Sistêmico , Linfócitos , Lúpus Eritematoso Sistêmico/imunologia , Humanos , Imunidade Inata/imunologia , Linfócitos/imunologia , Animais , Inflamação/imunologia , Inflamação/patologia
9.
Cell Immunol ; 399-400: 104824, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38615612

RESUMO

Infection of the respiratory tract with respiratory syncytial virus (RSV) is common and occurs repeatedly throughout life with most severe disease occurring at the extremes of age: in young infants and the elderly. Effective anti-viral therapeutics are not available and therefore prevention has been the primary strategy for reducing the disease burden. Our current understanding of respiratory mucosal cell biology and the immune response within the respiratory tract is inadequate to prevent infection caused by a pathogen like RSV that does not disseminate outside of this environment. Gaps in our understanding of the activation of innate and adaptive immunity in response to RSV and the role of age upon infection also limit improvements in the design of therapeutics and vaccines for young infants. However, advancements in structural biology have improved our ability to characterize antibodies against viral proteins and in 2023 the first vaccines for those over 60 years and pregnant women became available, potentially reducing the burden of disease. This review will examine our current understanding of the critical facets of anti-RSV immune responses in infants and young children as well as highlight areas where more research is needed.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Infecções por Vírus Respiratório Sincicial , Humanos , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Lactente , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Pré-Escolar , Anticorpos Antivirais/imunologia , Feminino , Vírus Sinciciais Respiratórios/imunologia
10.
Nature ; 628(8009): 844-853, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38570685

RESUMO

Mitochondria are critical modulators of antiviral tolerance through the release of mitochondrial RNA and DNA (mtDNA and mtRNA) fragments into the cytoplasm after infection, activating virus sensors and type-I interferon (IFN-I) response1-4. The relevance of these mechanisms for mitochondrial diseases remains understudied. Here we investigated mitochondrial recessive ataxia syndrome (MIRAS), which is caused by a common European founder mutation in DNA polymerase gamma (POLG1)5. Patients homozygous for the MIRAS variant p.W748S show exceptionally variable ages of onset and symptoms5, indicating that unknown modifying factors contribute to disease manifestation. We report that the mtDNA replicase POLG1 has a role in antiviral defence mechanisms to double-stranded DNA and positive-strand RNA virus infections (HSV-1, TBEV and SARS-CoV-2), and its p.W748S variant dampens innate immune responses. Our patient and knock-in mouse data show that p.W748S compromises mtDNA replisome stability, causing mtDNA depletion, aggravated by virus infection. Low mtDNA and mtRNA release into the cytoplasm and a slow IFN response in MIRAS offer viruses an early replicative advantage, leading to an augmented pro-inflammatory response, a subacute loss of GABAergic neurons and liver inflammation and necrosis. A population databank of around 300,000 Finnish individuals6 demonstrates enrichment of immunodeficient traits in carriers of the POLG1 p.W748S mutation. Our evidence suggests that POLG1 defects compromise antiviral tolerance, triggering epilepsy and liver disease. The finding has important implications for the mitochondrial disease spectrum, including epilepsy, ataxia and parkinsonism.


Assuntos
Alelos , DNA Polimerase gama , Vírus da Encefalite Transmitidos por Carrapatos , Herpesvirus Humano 1 , Tolerância Imunológica , SARS-CoV-2 , Animais , Feminino , Humanos , Masculino , Camundongos , Idade de Início , COVID-19/imunologia , COVID-19/virologia , COVID-19/genética , DNA Polimerase gama/genética , DNA Polimerase gama/imunologia , DNA Polimerase gama/metabolismo , DNA Mitocondrial/imunologia , DNA Mitocondrial/metabolismo , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/genética , Encefalite Transmitida por Carrapatos/imunologia , Encefalite Transmitida por Carrapatos/virologia , Efeito Fundador , Técnicas de Introdução de Genes , Herpes Simples/genética , Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/imunologia , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Imunidade Inata/genética , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Doenças Mitocondriais/enzimologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/imunologia , Mutação , RNA Mitocondrial/imunologia , RNA Mitocondrial/metabolismo , SARS-CoV-2/imunologia
11.
JCI Insight ; 9(8)2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38470489

RESUMO

Allergic airway disease (AAD) is an example of type 2 inflammation that leads to chronic airway eosinophilia controlled by CD4 Th2 cells. Inflammation is reinforced by mast cells and basophils armed with allergen-specific IgE made by allergen-specific B2 B cells of the adaptive immune system. Little is known about how AAD is affected by innate B1 cells, which produce natural antibodies (NAbs) that facilitate apoptotic cell clearance and detect damage- and pathogen-associated molecular patterns (DAMPS and PAMPS). We used transgenic mice lacking either B cells or NAbs in distinct mouse models of AAD that require either DAMPS or PAMPS as the initial trigger for type 2 immunity. In a DAMP-induced allergic model, driven by alum and uric acid, mouse strains lacking B cells (CD19DTA), NAbs (IgHEL MD4), or all secreted antibodies (sIgm-/-Aid-/-) displayed a significant reduction in both eosinophilia and Th2 priming compared with WT or Aid-/- mice lacking only germinal center-dependent high-affinity class-switched antibodies. Replenishing B cell-deficient mice with either unimmunized B1 B cells or NAbs during sensitization restored eosinophilia, suggesting that NAbs are required for licensing antigen-presenting cells to prime type 2 immunity. Conversely, PAMP-dependent type 2 priming to house dust mite or Aspergillus was not dependent on NAbs. This study reveals an underappreciated role of B1 B cell-generated NAbs in selectively driving DAMP-induced type 2 immunity.


Assuntos
Linfócitos B , Animais , Camundongos , Linfócitos B/imunologia , Células Th2/imunologia , Modelos Animais de Doenças , Camundongos Transgênicos , Camundongos Knockout , Imunidade Inata/imunologia , Camundongos Endogâmicos C57BL , Imunoglobulina E/imunologia , Alarminas/imunologia , Anticorpos/imunologia , Hipersensibilidade/imunologia , Eosinofilia/imunologia
12.
Int J Cancer ; 155(2): 352-364, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38483404

RESUMO

Treatment for higher-risk non-muscle invasive bladder cancer (NMIBC) involves intravesical immunotherapy with Bacillus Calmette Guérin (BCG); however, disease recurrence and progression occur frequently. Systemic immunity is critical for successful cancer immunotherapy; thus, recurrence of NMIBC may be due to suboptimal systemic activation of anti-tumor immunity after local immunotherapy. We previously reported that systemically acquired trained immunity (a form of innate immune memory) in circulating monocytes is associated with increased time-to-recurrence in patients with NMIBC treated with BCG. Herein, we used a mouse model of NMIBC to compare the effects of intravesical versus intravenous (systemic) BCG immunotherapy on the local and peripheral immune microenvironments. We also assessed whether BCG-induced trained immunity modulates anti-tumor immune responses. Compared with intravesical BCG, which led to a tumor-promoting immune microenvironment, intravenous BCG resulted in an anti-tumoral bladder microenvironment characterized by increased proportions of cytotoxic T lymphocytes (CTLs), and decreased proportions of myeloid-derived suppressor cells. Polarization toward anti-tumoral immunity occurred in draining lymph nodes, spleen, and bone marrow following intravenous versus intravesical BCG treatment. Pre-treatment with intravesical BCG was associated with increased rate of tumor growth compared with intravenous BCG pre-treatment. Trained immunity contributed to remodeling of the tumor immune microenvironment, as co-instillation of BCG-trained macrophages with ovalbumin-expressing bladder tumor cells increased the proportion of tumor-specific CTLs. Furthermore, BCG-trained dendritic cells exhibited enhanced antigen uptake and presentation and promoted CTL proliferation. Our data support the concept that systemic immune activation promotes anti-tumor responses, and that BCG-induced trained immunity is important in driving anti-tumor adaptive immunity.


Assuntos
Vacina BCG , Imunoterapia , Microambiente Tumoral , Neoplasias da Bexiga Urinária , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/terapia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Microambiente Tumoral/imunologia , Camundongos , Vacina BCG/imunologia , Vacina BCG/administração & dosagem , Vacina BCG/uso terapêutico , Imunoterapia/métodos , Feminino , Administração Intravesical , Camundongos Endogâmicos C57BL , Linfócitos T Citotóxicos/imunologia , Humanos , Modelos Animais de Doenças , Imunidade Inata/imunologia , Linhagem Celular Tumoral , Memória Imunológica/imunologia , Células Supressoras Mieloides/imunologia , Imunidade Treinada
13.
Eur J Immunol ; 54(5): e2350392, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38361213

RESUMO

Extracellular vesicles (EVs) are nanosized particles released by nearly every cell type across all kingdoms of life. As a result, EVs are ubiquitously present in various human body fluids. Composed of a lipid bilayer, EVs encapsulate proteins, nucleic acids, and metabolites, thus playing a crucial role in immunity, for example, by enabling intercellular communication. More recently, there has been increasing evidence that EVs can also act as key regulators of allergic immune responses. Their ability to facilitate cell-to-cell contact and to transport a variety of different biomolecules enables active modulation of both innate and adaptive immune processes associated with allergic reactions. A comprehensive understanding of the intricate mechanisms underlying the interactions among allergens, immune cells, and EVs is imperative to develop innovative strategies for controlling allergic responses. This review highlights the recent roles of host cell- and bacteria-derived EVs in allergic diseases, presenting experimental and clinical evidence that underscores their significance. Additionally, the therapeutic potential of EVs in allergy management is outlined, along with the challenges associated with targeted delivery and cargo stability for clinical use. Optimization of EV composition and targeting strategies holds promise for advancing translational applications and establishing EVs as biomarkers or safe therapeutics for assessing allergic reactions. For these reasons, EVs represent a promising avenue for advancing both our understanding and management of allergic immune processes.


Assuntos
Vesículas Extracelulares , Hipersensibilidade , Humanos , Vesículas Extracelulares/imunologia , Hipersensibilidade/imunologia , Hipersensibilidade/terapia , Animais , Comunicação Celular/imunologia , Imunidade Inata/imunologia , Alérgenos/imunologia , Imunidade Adaptativa/imunologia , Biomarcadores
14.
J Immunol ; 212(2): 335-345, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38047899

RESUMO

Although electric field-induced cell membrane permeabilization (electroporation) is used in a wide range of clinical applications from cancer therapy to cardiac ablation, the cellular- and molecular-level details of the processes that determine the success or failure of these treatments are poorly understood. Nanosecond pulsed electric field (nsPEF)-based tumor therapies are known to have an immune component, but whether and how immune cells sense the electroporative damage and respond to it have not been demonstrated. Damage- and pathogen-associated stresses drive inflammation via activation of cytosolic multiprotein platforms known as inflammasomes. The assembly of inflammasome complexes triggers caspase-1-dependent secretion of IL-1ß and in many settings a form of cell death called pyroptosis. In this study we tested the hypothesis that the nsPEF damage is sensed intracellularly by the NLRP3 inflammasome. We found that 200-ns PEFs induced aggregation of the inflammasome adaptor protein ASC, activation of caspase-1, and triggered IL-1ß release in multiple innate immune cell types (J774A.1 macrophages, bone marrow-derived macrophages, and dendritic cells) and in vivo in mouse skin. Efflux of potassium from the permeabilized cell plasma membrane was partially responsible for nsPEF-induced inflammasome activation. Based on results from experiments using both the NRLP3-specific inhibitor MCC950 and NLRP3 knockout cells, we propose that the damage created by nsPEFs generates a set of stimuli for the inflammasome and that more than one sensor can drive IL-1ß release in response to electrical pulse stimulation. This study shows, to our knowledge, for the first time, that PEFs activate the inflammasome, suggesting that this pathway alarms the immune system after treatment.


Assuntos
Inflamassomos , Interleucina-1beta , Macrófagos , Pele , Inflamassomos/imunologia , Interleucina-1beta/imunologia , Animais , Camundongos , Pele/imunologia , Células HEK293 , Humanos , Linhagem Celular , Gasderminas/imunologia , Estimulação Elétrica , Macrófagos/imunologia , Imunidade Inata/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
15.
J Virol ; 97(12): e0150123, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37982618

RESUMO

IMPORTANCE: The type-I interferon (IFN-I) signaling pathway is the first line of antiviral innate immunity. It must be precisely regulated against virus-induced damage. The tightly regulated mechanisms of action of host genes in the antiviral innate immune signaling pathway are still worth studying. Here, we report a novel role of DLG1 in positively regulating the IκB kinase epsilon (IKKε)-mediated IFN-I signaling response against negative-stranded RNA virus replication, whereas the RNA virus inhibits the expression of DLG1 for immune escape. Importantly, the E3 ligase March2 interacts with and promotes K27-linked polyubiquitination of IKKε, and p62 is a cargo receptor that recognizes ubiquitinated IKKε for eventual autophagic degradation. Together, the current findings elucidate the role of DLG1 in the antiviral IFN-I signaling pathway and viral infection repression.


Assuntos
Autofagia , Proteína 1 Homóloga a Discs-Large , Quinase I-kappa B , Imunidade Inata , Vírus de RNA de Sentido Negativo , Proteína Sequestossoma-1 , Viroses , Humanos , Proteína 1 Homóloga a Discs-Large/metabolismo , Quinase I-kappa B/metabolismo , Imunidade Inata/imunologia , Vírus de RNA de Sentido Negativo/crescimento & desenvolvimento , Vírus de RNA de Sentido Negativo/imunologia , Poliubiquitina/metabolismo , Proteína Sequestossoma-1/antagonistas & inibidores , Transdução de Sinais , Viroses/imunologia , Animais , Linhagem Celular
16.
J Virol ; 97(12): e0130423, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37982645

RESUMO

IMPORTANCE: Interferon-stimulated genes (ISGs) are induced in response to interferon expression due to viral infections. Role of these ISGs can be variable in different cells or organs. Our study highlights such cell-specific role of an ISG, Ddx3, which regulates the translation of mRNAs essential for interferon induction (PACT) and interferon signaling (STAT1) in a cell-specific manner. Our study also highlights the role of PACT in RNA virus-induced RLR signaling. Our study depicts how Ddx3 regulates innate immune signaling pathways in an indirect manner. Such cell-specific behavior of ISGs helps us to better understand viral pathogenesis and highlights the complexities of viral tropism and innate immune responses.


Assuntos
Imunidade Inata , Interferons , Vírus de RNA , Imunidade Inata/imunologia , Interferons/biossíntese , Interferons/imunologia , Vírus de RNA/imunologia , Vírus de RNA/patogenicidade , Transdução de Sinais , Humanos , Animais , Camundongos
17.
N Engl J Med ; 389(13): 1203-1210, 2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37754285

RESUMO

We treated a 27-year-old patient with Duchenne's muscular dystrophy (DMD) with recombinant adeno-associated virus (rAAV) serotype 9 containing dSaCas9 (i.e., "dead" Staphylococcus aureus Cas9, in which the Cas9 nuclease activity has been inactivated) fused to VP64; this transgene was designed to up-regulate cortical dystrophin as a custom CRISPR-transactivator therapy. The dose of rAAV used was 1×1014 vector genomes per kilogram of body weight. Mild cardiac dysfunction and pericardial effusion developed, followed by acute respiratory distress syndrome (ARDS) and cardiac arrest 6 days after transgene treatment; the patient died 2 days later. A postmortem examination showed severe diffuse alveolar damage. Expression of transgene in the liver was minimal, and there was no evidence of AAV serotype 9 antibodies or effector T-cell reactivity in the organs. These findings indicate that an innate immune reaction caused ARDS in a patient with advanced DMD treated with high-dose rAAV gene therapy. (Funded by Cure Rare Disease.).


Assuntos
Distrofina , Terapia Genética , Distrofia Muscular de Duchenne , Síndrome do Desconforto Respiratório , Transgenes , Adulto , Humanos , Anticorpos , Distrofina/genética , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/imunologia , Transgenes/genética , Transgenes/imunologia , Evolução Fatal , Imunidade Inata/genética , Imunidade Inata/imunologia
18.
Nat Immunol ; 24(8): 1256-1264, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37400674

RESUMO

Innate lymphoid cells (ILCs) can quickly switch from a quiescent state to an active state and rapidly produce effector molecules that provide critical early immune protection. How the post-transcriptional machinery processes different stimuli and initiates robust gene expression in ILCs is poorly understood. Here, we show that deletion of the N6-methyladenosine (m6A) writer protein METTL3 has little impact on ILC homeostasis or cytokine-induced ILC1 or ILC3 responses but significantly diminishes ILC2 proliferation, migration and effector cytokine production and results in impaired antihelminth immunity. m6A RNA modification supports an increase in cell size and transcriptional activity in activated ILC2s but not in ILC1s or ILC3s. Among other transcripts, the gene encoding the transcription factor GATA3 is highly m6A methylated in ILC2s. Targeted m6A demethylation destabilizes nascent Gata3 mRNA and abolishes the upregulation of GATA3 and ILC2 activation. Our study suggests a lineage-specific requirement of m6A for ILC2 responses.


Assuntos
Imunidade Inata , Linfócitos , Citocinas/metabolismo , Homeostase , Imunidade Inata/genética , Imunidade Inata/imunologia , Linfócitos/imunologia , RNA/metabolismo , Animais , Camundongos
19.
Int J Mol Sci ; 24(13)2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37446157

RESUMO

Atherosclerotic cardiovascular disease is the most common cause of morbidity and death worldwide. Recent studies have demonstrated that this chronic inflammatory disease of the arterial wall can be controlled through the modulation of immune system activity. Many patients with cardiovascular disease remain at elevated risk of recurrent events despite receiving current, state-of-the-art preventive medical treatment. Much of this residual risk is attributed to inflammation. Therefore, finding new treatment strategies for this category of patients became of common interest. This review will discuss the experimental and clinical data supporting the possibility of developing immune-based therapies for lowering cardiovascular risk, explicitly focusing on vaccination strategies.


Assuntos
Aterosclerose , Imunomodulação , Humanos , Aterosclerose/imunologia , Aterosclerose/prevenção & controle , Aterosclerose/terapia , Fatores de Risco de Doenças Cardíacas , Inflamação , Vacinação/tendências , Imunidade Inata/imunologia , Imunidade Adaptativa/imunologia , Imunidade Humoral/imunologia , Autoantígenos/imunologia , Ensaios Clínicos como Assunto , Vacinas/imunologia , Vacinas/uso terapêutico
20.
Nature ; 620(7974): 634-642, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37438525

RESUMO

The physiological functions of mast cells remain largely an enigma. In the context of barrier damage, mast cells are integrated in type 2 immunity and, together with immunoglobulin E (IgE), promote allergic diseases. Allergic symptoms may, however, facilitate expulsion of allergens, toxins and parasites and trigger future antigen avoidance1-3. Here, we show that antigen-specific avoidance behaviour in inbred mice4,5 is critically dependent on mast cells; hence, we identify the immunological sensor cell linking antigen recognition to avoidance behaviour. Avoidance prevented antigen-driven adaptive, innate and mucosal immune activation and inflammation in the stomach and small intestine. Avoidance was IgE dependent, promoted by Th2 cytokines in the immunization phase and by IgE in the execution phase. Mucosal mast cells lining the stomach and small intestine rapidly sensed antigen ingestion. We interrogated potential signalling routes between mast cells and the brain using mutant mice, pharmacological inhibition, neural activity recordings and vagotomy. Inhibition of leukotriene synthesis impaired avoidance, but overall no single pathway interruption completely abrogated avoidance, indicating complex regulation. Collectively, the stage for antigen avoidance is set when adaptive immunity equips mast cells with IgE as a telltale of past immune responses. On subsequent antigen ingestion, mast cells signal termination of antigen intake. Prevention of immunopathology-causing, continuous and futile responses against per se innocuous antigens or of repeated ingestion of toxins through mast-cell-mediated antigen-avoidance behaviour may be an important arm of immunity.


Assuntos
Alérgenos , Aprendizagem da Esquiva , Hipersensibilidade , Mastócitos , Animais , Camundongos , Alérgenos/imunologia , Aprendizagem da Esquiva/fisiologia , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Mastócitos/imunologia , Estômago/imunologia , Vagotomia , Imunidade Inata/imunologia , Imunidade nas Mucosas/imunologia , Células Th2/imunologia , Citocinas/imunologia , Leucotrienos/biossíntese , Leucotrienos/imunologia , Intestino Delgado/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA