Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-36675299

RESUMO

It is well ascertained that airway inflammation has a key role in the genesis of numerous respiratory pathologies, including asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Pulmonary tissue inflammation and anti-inflammatory responses implicate an intricate relationship between local and infiltrating immune cells and structural pulmonary cells. Alarmins are endogenic proteins discharged after cell injury in the extracellular microenvironment. The purpose of our review is to highlight the alterations in respiratory diseases involving some alarmins, such as high mobility group box 1 (HMGB1) and interleukin (IL)-33, and their inter-relationships and relationships with genetic non-coding material, such as microRNAs. The role played by these alarmins in some pathophysiological processes confirms the existence of an axis composed of HMGB1 and IL-33. These alarmins have been implicated in ferroptosis, the onset of type 2 inflammation and airway alterations. Moreover, both factors can act on non-coding genetic material capable of modifying respiratory function. Finally, we present an outline of alarmins and RNA-based therapeutics that have been proposed to treat respiratory pathologies.


Assuntos
Proteína HMGB1 , MicroRNAs , Transtornos Respiratórios , Humanos , Alarminas/genética , MicroRNAs/genética , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Inflamação/genética , Inflamação/terapia
2.
Biochim Biophys Acta Mol Cell Res ; 1870(2): 119408, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36503009

RESUMO

Upon microbial infections with the subsequent host response of innate immunity, a variety of fragmented RNA- and DNA-based "Pathogen-associated molecular patterns" (PAMPs) are recognized mainly by endosomal or cytoplasmic host cell "Pattern recognition receptors" (PRRs), particularly "Toll-like receptors" (TLRs). Concomitantly, various self-extracellular RNA species (exRNAs) are present in extracellular body fluids where they contribute to diverse physiological and homeostatic processes. In principle, such exRNAs, including the most abundant one, ribosomal exRNA (rexRNA), are designated as "Danger-associated molecular patterns" (DAMPs) and are prevented by e.g. natural modifications from uncontrolled signaling via TLRs to avoid hyper-inflammatory responses or autoimmunity. Upon cellular stress or tissue damage/necrosis, the levels and composition of released self-exRNA species, either in free form, in complex with proteins or in association with extracellular vesicles (EVs), can change considerably. Among the self-exRNAs, rexRNA is considered as a non-typical DAMP, since it may induce inflammatory responses by cell membrane receptors, both in the absence or presence of PAMPs. Yet, its mode of receptor activation to mount inflammatory responses remains obscure. RexRNA also serves as a universal damaging factor in cardiovascular and other diseases independent of PRRs. In general, RNase1 provides a profound antagonist in these pathologies and in rexRNA-mediated inflammatory cell responses. Based on the extrapolation of the here described aspects of rexRNA-biology, further activities of this molecular entity are hypothesized that may stimulate additional research in this area.


Assuntos
Moléculas com Motivos Associados a Patógenos , RNA Ribossômico , RNA Ribossômico/genética , Imunidade Inata , Receptores de Reconhecimento de Padrão/metabolismo , RNA/genética , Alarminas/genética
3.
Int J Mol Sci ; 23(19)2022 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-36233010

RESUMO

A generally accepted hypothesis for the initial activation of an immune or autoimmune response argues that alarmins are released from injured, dying and/or activated immune cells, and these products complex with receptors that activate signal transduction pathways and recruit immune cells to the site of injury where the recruited cells are stimulated to initiate immune and/or cellular repair responses. While there are multiple diverse families of alarmins such as interleukins (IL), heat-shock proteins (HSP), Toll-like receptors (TLR), plus individual molecular entities such as Galectin-3, Calreticulin, Thymosin, alpha-Defensin-1, RAGE, and Interferon-1, one phylogenetically conserved family are the Annexin proteins known to promote an extensive range of biomolecular and cellular products that can directly and indirectly regulate inflammation and immune activities. For the present report, we examined the temporal expression profiles of the 12 mammalian annexin genes (Anxa1-11 and Anxa13), applying our temporal genome-wide transcriptome analyses of ex vivo salivary and lacrimal glands from our C57BL/6.NOD-Aec1Aec2 mouse model of Sjögren's Syndrome (SS), a human autoimmune disease characterized primarily by severe dry mouth and dry eye symptoms. Results indicate that annexin genes Anax1-7 and -11 exhibited upregulated expressions and the initial timing for these upregulations occurred as early as 8 weeks of age and prior to any covert signs of a SS-like disease. While the profiles of the two glands were similar, they were not identical, suggesting the possibility that the SS-like disease may not be uniform in the two glands. Nevertheless, this early pre-clinical and concomitant upregulated expression of this specific set of alarmins within the immune-targeted organs represents a potential target for identifying the pre-clinical stage in human SS as well, a fact that would clearly impact future interventions and therapeutic strategies.


Assuntos
Anexinas , Aparelho Lacrimal , Síndrome de Sjogren , Timosina , Alarminas/genética , Alarminas/metabolismo , Animais , Anexinas/genética , Anexinas/metabolismo , Calreticulina/metabolismo , Modelos Animais de Doenças , Galectina 3/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Interferons/metabolismo , Aparelho Lacrimal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , RNA/metabolismo , Timosina/genética , Transcriptoma , alfa-Defensinas/genética
4.
Proc Natl Acad Sci U S A ; 119(36): e2120680119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-35998224

RESUMO

The systemic immune response to viral infection is shaped by master transcription factors, such as NF-κB, STAT1, or PU.1. Although long noncoding RNAs (lncRNAs) have been suggested as important regulators of transcription factor activity, their contributions to the systemic immunopathologies observed during SARS-CoV-2 infection have remained unknown. Here, we employed a targeted single-cell RNA sequencing approach to reveal lncRNAs differentially expressed in blood leukocytes during severe COVID-19. Our results uncover the lncRNA PIRAT (PU.1-induced regulator of alarmin transcription) as a major PU.1 feedback-regulator in monocytes, governing the production of the alarmins S100A8/A9, key drivers of COVID-19 pathogenesis. Knockout and transgene expression, combined with chromatin-occupancy profiling, characterized PIRAT as a nuclear decoy RNA, keeping PU.1 from binding to alarmin promoters and promoting its binding to pseudogenes in naïve monocytes. NF-κB-dependent PIRAT down-regulation during COVID-19 consequently releases a transcriptional brake, fueling alarmin production. Alarmin expression is additionally enhanced by the up-regulation of the lncRNA LUCAT1, which promotes NF-κB-dependent gene expression at the expense of targets of the JAK-STAT pathway. Our results suggest a major role of nuclear noncoding RNA networks in systemic antiviral responses to SARS-CoV-2 in humans.


Assuntos
COVID-19 , Regulação da Expressão Gênica , Monócitos , RNA Longo não Codificante , SARS-CoV-2 , Alarminas/genética , COVID-19/genética , COVID-19/imunologia , Humanos , Janus Quinases/genética , Monócitos/imunologia , NF-kappa B/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA-Seq , SARS-CoV-2/imunologia , Fatores de Transcrição STAT/genética , Transdução de Sinais/genética , Análise de Célula Única
5.
Sci Rep ; 12(1): 2814, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35181730

RESUMO

Non-apoptotic regulated cell death (ferroptosis and necroptosis) leads to the release of damage-associated molecular patterns (DAMPs), which initiate and perpetuate a non-infectious inflammatory response. We hypothesize that DAMPs and non-apoptotic regulated cell death are critical players of atherosclerotic plaque progression with inadequate response to lipid-lowering treatment. We aimed to uncover the silent mechanisms that govern the existing residual risk of cardiovascular-related mortality in experimental atherosclerosis. Proteomic and genomic approaches were applied on the ascending aorta of hyperlipidemic rabbits and controls with and without lipid-lowering treatment. The hyperlipidemic animals, which presented numerous heterogeneous atherosclerotic lesions, exhibited high concentrations of serum lipids and increased lipid peroxidation oxidative stress markers. The analyses revealed the significant upregulation of DAMPs and proteins implicated in ferroptosis and necroptosis by hyperlipidemia. Some of them did not respond to lipid-lowering treatment. Dysregulation of five proteins involved in non-apoptotic regulated cell death proteins (VDAC1, VDAC3, FTL, TF and PCBP1) and nine associated DAMPs (HSP90AA1, HSP90AB1, ANXA1, LGALS3, HSP90B1, S100A11, FN, CALR, H3-3A) was not corrected by the treatment. These proteins could play a key role in the atherosclerotic silent evolution and may possess an unexplored therapeutic potential. Mass spectrometry data are available via ProteomeXchange with identifier PXD026379.


Assuntos
Alarminas/genética , Aterosclerose/genética , Lipídeos/sangue , Placa Aterosclerótica/genética , Alarminas/sangue , Animais , Aorta/metabolismo , Aorta/patologia , Apoptose/genética , Aterosclerose/sangue , Aterosclerose/patologia , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica/genética , Humanos , Peroxidação de Lipídeos/genética , Lipídeos/genética , Espectrometria de Massas , Estresse Oxidativo/genética , Placa Aterosclerótica/sangue , Placa Aterosclerótica/patologia , Proteoma/metabolismo , Coelhos
6.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34830328

RESUMO

Epidermolysis bullosa simplex (EBS) is a group of inherited keratinopathies that, in most cases, arise due to mutations in keratins and lead to intraepidermal ruptures. The cellular pathology of most EBS subtypes is associated with the fragility of the intermediate filament network, cytolysis of the basal layer of the epidermis, or attenuation of hemidesmosomal/desmosomal components. Mutations in keratins 5/14 or in other genes that encode associated proteins induce structural disarrangements of different strengths depending on their locations in the genes. Keratin aggregates display impaired dynamics of assembly and diminished solubility and appear to be the trigger for endoplasmic reticulum (ER) stress upon being phosphorylated by MAPKs. Global changes in cellular signaling mainly occur in cases of severe dominant EBS mutations. The spectrum of changes initiated by phosphorylation includes the inhibition of proteasome degradation, TNF-α signaling activation, deregulated proliferation, abnormal cell migration, and impaired adherence of keratinocytes. ER stress also leads to the release of proinflammatory danger-associated molecular pattern (DAMP) molecules, which enhance avalanche-like inflammation. Many instances of positive feedback in the course of cellular stress and the development of sterile inflammation led to systemic chronic inflammation in EBS. This highlights the role of keratin in the maintenance of epidermal and immune homeostasis.


Assuntos
Alarminas/genética , Epiderme/metabolismo , Epidermólise Bolhosa Simples/genética , Queratina-14/genética , Queratina-5/genética , Queratinócitos/metabolismo , Alarminas/metabolismo , Estresse do Retículo Endoplasmático/genética , Epiderme/patologia , Epidermólise Bolhosa Simples/metabolismo , Epidermólise Bolhosa Simples/patologia , Regulação da Expressão Gênica , Humanos , Inflamação , Filamentos Intermediários/metabolismo , Filamentos Intermediários/patologia , Filamentos Intermediários/ultraestrutura , Queratina-14/metabolismo , Queratina-5/metabolismo , Queratinócitos/patologia , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Complexo de Endopeptidases do Proteassoma/metabolismo , Agregados Proteicos , Proteólise , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
Nat Immunol ; 22(8): 947-957, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34239121

RESUMO

One of most challenging issues in tumor immunology is a better understanding of the dynamics in the accumulation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TIME), as this would lead to the development of new cancer therapeutics. Here, we show that translationally controlled tumor protein (TCTP) released by dying tumor cells is an immunomodulator crucial to full-blown MDSC accumulation in the TIME. We provide evidence that extracellular TCTP mediates recruitment of the polymorphonuclear MDSC (PMN-MDSC) population in the TIME via activation of Toll-like receptor-2. As further proof of principle, we show that inhibition of TCTP suppresses PMN-MDSC accumulation and tumor growth. In human cancers, we find an elevation of TCTP and an inverse correlation of TCTP gene dosage with antitumor immune signatures and clinical prognosis. This study reveals the hitherto poorly understood mechanism of the MDSC dynamics in the TIME, offering a new rationale for cancer immunotherapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Quimiocina CXCL1/metabolismo , Neoplasias Colorretais/imunologia , Células Supressoras Mieloides/imunologia , Receptor 2 Toll-Like/imunologia , Microambiente Tumoral/imunologia , Alarminas/genética , Alarminas/metabolismo , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células RAW 264.7 , Proteína Tumoral 1 Controlada por Tradução
8.
Int J Mol Sci ; 22(14)2021 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-34299310

RESUMO

Oxidative stress is a major contributor to the pathogenesis of various inflammatory diseases. Accumulating evidence has shown that oxidative stress is characterized by the overproduction of reactive oxygen species (ROS). Previous reviews have highlighted inflammatory signaling pathways, biomarkers, molecular targets, and pathogenetic functions mediated by oxidative stress in various diseases. The inflammatory signaling cascades are initiated through the recognition of host cell-derived damage associated molecular patterns (DAMPs) and microorganism-derived pathogen associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs). In this review, the effects of PRRs from the Toll-like (TLRs), the retinoic acid-induced gene I (RIG-I)-like receptors (RLRs) and the NOD-like (NLRs) families, and the activation of these signaling pathways in regulating the production of ROS and/or oxidative stress are summarized. Furthermore, important directions for future studies, especially for pathogen-induced signaling pathways through oxidative stress are also reviewed. The present review will highlight potential therapeutic strategies relevant to inflammatory diseases based on the correlations between ROS regulation and PRRs-mediated signaling pathways.


Assuntos
Inflamação/metabolismo , Receptores de Reconhecimento de Padrão/metabolismo , Alarminas/genética , Alarminas/imunologia , Alarminas/metabolismo , Animais , Autofagia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/terapia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Proteína DEAD-box 58/metabolismo , Interações entre Hospedeiro e Microrganismos , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Inflamassomos/metabolismo , Inflamação/genética , Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/terapia , Modelos Biológicos , Proteínas NLR/genética , Proteínas NLR/imunologia , Proteínas NLR/metabolismo , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/terapia , Estresse Oxidativo , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Ativação Transcricional
9.
EMBO J ; 40(16): e106540, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34121210

RESUMO

Dendritic cells (DC) subsets, like Langerhans cells (LC), are immune cells involved in pathogen sensing. They express specific antimicrobial cellular factors that are able to restrict infection and limit further pathogen transmission. Here, we identify the alarmin S100A9 as a novel intracellular antiretroviral factor expressed in human monocyte-derived and skin-derived LC. The intracellular expression of S100A9 is decreased upon LC maturation and inversely correlates with enhanced susceptibility to HIV-1 infection of LC. Furthermore, silencing of S100A9 in primary human LC relieves HIV-1 restriction while ectopic expression of S100A9 in various cell lines promotes intrinsic resistance to both HIV-1 and MLV infection by acting on reverse transcription. Mechanistically, the intracellular expression of S100A9 alters viral capsid uncoating and reverse transcription. S100A9 also shows potent inhibitory effect against HIV-1 and MMLV reverse transcriptase (RTase) activity in vitro in a divalent cation-dependent manner. Our findings uncover an unexpected intracellular function of the human alarmin S100A9 in regulating antiretroviral immunity in Langerhans cells.


Assuntos
Alarminas/genética , Calgranulina B/genética , HIV-1/fisiologia , Células de Langerhans/virologia , Vírus da Leucemia Murina de Moloney/fisiologia , Infecções por Retroviridae/prevenção & controle , Animais , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Cricetulus , HIV-1/genética , Interações Hospedeiro-Patógeno , Humanos , Células de Langerhans/imunologia , Leucemia Experimental/prevenção & controle , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Transcrição Reversa , Fator de Crescimento Transformador beta/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Replicação Viral
10.
Cells ; 10(3)2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33802550

RESUMO

Mitochondria serve as a hub for a multitude of vital cellular processes. To ensure an efficient deployment of mitochondrial tasks, organelle homeostasis needs to be preserved. Mitochondrial quality control (MQC) mechanisms (i.e., mitochondrial dynamics, biogenesis, proteostasis, and autophagy) are in place to safeguard organelle integrity and functionality. Defective MQC has been reported in several conditions characterized by chronic low-grade inflammation. In this context, the displacement of mitochondrial components, including mitochondrial DNA (mtDNA), into the extracellular compartment is a possible factor eliciting an innate immune response. The presence of bacterial-like CpG islands in mtDNA makes this molecule recognized as a damaged-associated molecular pattern by the innate immune system. Following cell death-triggering stressors, mtDNA can be released from the cell and ignite inflammation via several pathways. Crosstalk between autophagy and apoptosis has emerged as a pivotal factor for the regulation of mtDNA release, cell's fate, and inflammation. The repression of mtDNA-mediated interferon production, a powerful driver of immunological cell death, is also regulated by autophagy-apoptosis crosstalk. Interferon production during mtDNA-mediated inflammation may be exploited for the elimination of dying cells and their conversion into elements driving anti-tumor immunity.


Assuntos
Apoptose/genética , DNA Mitocondrial/genética , Inflamação/genética , Mitocôndrias/genética , Mitofagia/genética , Neoplasias/genética , Alarminas/genética , Alarminas/imunologia , Apoptose/imunologia , DNA Mitocondrial/imunologia , Regulação da Expressão Gênica , Homeostase/genética , Homeostase/imunologia , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/patologia , Interferons/genética , Interferons/imunologia , Mitocôndrias/imunologia , Mitocôndrias/patologia , Dinâmica Mitocondrial/genética , Dinâmica Mitocondrial/imunologia , Mitofagia/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Estresse Oxidativo , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
11.
Front Immunol ; 12: 599296, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33717078

RESUMO

We have demonstrated that intranasal immunotherapy with allergens formulated in a nanoemulsion (NE) mucosal adjuvant suppresses Th2/IgE-mediated allergic responses and protects from allergen challenge in murine food allergy models. Protection conferred by this therapy is associated with strong suppression of allergen specific Th2 cellular immunity and increased Th1 cytokines. Here we extend these studies to examine the effect of NE-allergen immunization in mice sensitized to multiple foods. Mice were sensitized to both egg and peanut and then received NE vaccine formulated with either one or both of these allergens. The animals were then subjected to oral challenges with either egg or peanut to assess reactivity. Immunization with NE formulations containing both egg and peanut markedly reduced reactivity after oral allergen challenge with either allergen. Interestingly, mice that received the vaccine containing only peanut also had reduced reactivity to challenge with egg. Protection from oral allergen challenge was achieved despite the persistence of allergen-specific IgE and was associated with strong suppression of both Th2-polarized immune responses, alarmins and type 2 innate lymphoid cells (ILC2). NE-induced bystander suppression of reactivity required IFN-γ and the presence of an allergen in the NE vaccine. These results demonstrate that anaphylactic reactions to food allergens can be suppressed using allergen-specific immunotherapy without having to eliminate allergen-specific IgE and suggests that modulation of Th2 immunity towards one allergen may induce bystander effects that suppress reactivity to other allergens through the induction of IFN-γ and suppression of alarmins in the intestine. In addition, these data suggest that a NE vaccine for a single food allergen may lead to a global suppression of allergic responses to multiple foods.


Assuntos
Alarminas/genética , Alérgenos/imunologia , Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/terapia , Regulação da Expressão Gênica , Vacinas/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Efeito Espectador , Citocinas/metabolismo , Dessensibilização Imunológica , Modelos Animais de Doenças , Imunoglobulina E/imunologia , Imunomodulação , Camundongos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Vacinas/administração & dosagem
12.
Cell Rep Med ; 2(3): 100209, 2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33763654

RESUMO

Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control.


Assuntos
Vacinas Anticâncer/administração & dosagem , Imunoterapia/métodos , Vírus da Coriomeningite Linfocítica/imunologia , Mastocitoma/terapia , Vírus Pichinde/imunologia , Linfócitos T Citotóxicos/imunologia , Alarminas/genética , Alarminas/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Feminino , Expressão Gênica , Engenharia Genética/métodos , Vetores Genéticos/classificação , Vetores Genéticos/imunologia , Cobaias , Imunização Secundária , Vírus da Coriomeningite Linfocítica/classificação , Vírus da Coriomeningite Linfocítica/genética , Mastocitoma/genética , Mastocitoma/imunologia , Mastocitoma/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Filogenia , Vírus Pichinde/classificação , Vírus Pichinde/genética , Tolerância a Antígenos Próprios , Análise de Sobrevida , Vacinação/métodos
13.
J Mol Med (Berl) ; 99(6): 729-741, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33666676

RESUMO

Chronic kidney disease is a widespread terminal illness that afflicts millions of people across the world. Hemodialysis is the predominant therapeutic management strategy for kidney failure and involves the external filtration of metabolic waste within the circulation. This process requires an arteriovenous fistula (AVF) for vascular access. However, AVF maturation failures are significant obstacles in establishing long-term vascular access for hemodialysis. Appropriate stimulation, activation, and proliferation of smooth muscle cells, proper endothelial cell orientation, adequate structural changes in the ECM, and the release of anti-inflammatory markers are associated with maturation. AVFs often fail to mature due to inadequate tissue repair and remodeling, leading to neointimal hyperplasia lesions. The transdifferentiation of myofibroblasts and sterile inflammation are possibly involved in AVF maturation failures; however, limited data is available in this regard. The present article critically reviews the interplay of various damage-associated molecular patterns (DAMPs) and the downstream sterile inflammatory signaling with a focus on the NLRP3 inflammasome. Improved knowledge concerning AVF maturation pathways can be unveiled by investigating the novel DAMPs and the mediators of sterile inflammation in vascular remodeling that would open improved therapeutic opportunities in the management of AVF maturation failures and its associated complications.


Assuntos
Fístula Arteriovenosa/etiologia , Suscetibilidade a Doenças , Inflamação/complicações , Alarminas/genética , Alarminas/metabolismo , Animais , Fístula Arteriovenosa/metabolismo , Biomarcadores , Humanos , Inflamação/etiologia , Transdução de Sinais , Receptores Toll-Like/metabolismo
14.
Front Immunol ; 12: 606622, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732235

RESUMO

Damage-associated molecular patterns (DAMPs) are a group of immunostimulatory molecules, which take part in inflammatory response after tissue injury. Kidney-specific DAMPs include Tamm-Horsfall glycoprotein, crystals, and uromodulin, released by tubular damage for example. Non-kidney-specific DAMPs include intracellular particles such as nucleus [histones, high-mobility group box 1 protein (HMGB1)] and cytosol parts. DAMPs trigger innate immunity by activating the NRLP3 inflammasome, G-protein coupled class receptors or the Toll-like receptor. Tubular necrosis leads to acute kidney injury (AKI) in either septic, ischemic or toxic conditions. Tubular necrosis releases DAMPs such as histones and HMGB1 and increases vascular permeability, which perpetuates shock and hypoperfusion via Toll Like Receptors. In acute tubular necrosis, intracellular abundance of NADPH may explain a chain reaction where necrosis spreads from cell to cell. The nature AKI in intensive care units does not have preclinical models that meet a variation of blood perfusion or a variation of glomerular filtration within hours before catecholamine infusion. However, the dampening of several DAMPs in AKI could provide organ protection. Research should be focused on the numerous pathophysiological pathways to identify the relative contribution to renal dysfunction. The therapeutic perspectives could be strategies to suppress side effect of DAMPs and to promote renal function regeneration.


Assuntos
Injúria Renal Aguda/etiologia , Injúria Renal Aguda/metabolismo , Alarminas/genética , Alarminas/metabolismo , Suscetibilidade a Doenças , Sepse/complicações , Injúria Renal Aguda/diagnóstico , Injúria Renal Aguda/terapia , Biomarcadores , Citocinas/metabolismo , Gerenciamento Clínico , Humanos , Testes de Função Renal , Recuperação de Função Fisiológica , Regeneração
15.
PLoS One ; 16(3): e0248996, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33780491

RESUMO

One of the central challenges for cancer therapy is the identification of factors in the tumor microenvironment that increase tumor progression and prevent immune surveillance. One such element associated with breast cancer is stromal fibrosis, a histopathologic criterion for invasive cancer and poor survival. Fibrosis is caused by inflammatory factors and remodeling of the extracellular matrix that elicit an immune tolerant microenvironment. To address the role of fibrosis in tumorigenesis, we developed NeuT/ATTAC transgenic mice expressing a constitutively active NeuT/erbB2 transgene, and an inducible, fat-directed caspase-8 fusion protein, which upon activation results in selective and partial ablation of mammary fat and its replacement with fibrotic tissue. Induction of fibrosis in NeuT/ATTAC mice led to more rapid tumor development and an inflammatory and fibrotic stromal environment. In an effort to explore therapeutic options that could reduce fibrosis and immune tolerance, mice were treated with the oxysterol liver X receptor (LXR) pan agonist, N,N-dimethyl-3-ß-hydroxy-cholenamide (DMHCA), an agent known to reduce fibrosis in non-malignant diseases. DMHCA reduced tumor progression, tumor multiplicity and fibrosis, and improved immune surveillance by reducing infiltrating myeloid-derived suppressor cells and increasing CD4 and CD8 effector T cells. These effects were associated with downregulation of an LXR-dependent gene network related to reduced breast cancer survival that included Spp1, S100a9, Anxa1, Mfge8 and Cd14. These findings suggest that the use of DMHCA may be a potentially effective approach to reduce desmoplasia and immune tolerance and increase the efficacy of cancer therapy.


Assuntos
Carcinogênese/imunologia , Carcinogênese/patologia , Receptores X do Fígado/agonistas , Receptor ErbB-2/metabolismo , Alarminas/genética , Alarminas/metabolismo , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Carcinogênese/efeitos dos fármacos , Ácidos Cólicos/farmacologia , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Fibrose , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores X do Fígado/metabolismo , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Células Supressoras Mieloides/efeitos dos fármacos , Células Supressoras Mieloides/metabolismo , Proteínas de Neoplasias/metabolismo
16.
Front Immunol ; 12: 611910, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33708206

RESUMO

Detrimental inflammatory responses after solid organ transplantation are initiated when immune cells sense pathogen-associated molecular patterns (PAMPs) and certain damage-associated molecular patterns (DAMPs) released or exposed during transplant-associated processes, such as ischemia/reperfusion injury (IRI), surgical trauma, and recipient conditioning. These inflammatory responses initiate and propagate anti-alloantigen (AlloAg) responses and targeting DAMPs and PAMPs, or the signaling cascades they activate, reduce alloimmunity, and contribute to improved outcomes after allogeneic solid organ transplantation in experimental studies. However, DAMPs have also been implicated in initiating essential anti-inflammatory and reparative functions of specific immune cells, particularly Treg and macrophages. Interestingly, DAMP signaling is also involved in local and systemic homeostasis. Herein, we describe the emerging literature defining how poor outcomes after transplantation may result, not from just an over-abundance of DAMP-driven inflammation, but instead an inadequate presence of a subset of DAMPs or related molecules needed to repair tissue successfully or re-establish tissue homeostasis. Adverse outcomes may also arise when these homeostatic or reparative signals become dysregulated or hijacked by alloreactive immune cells in transplant niches. A complete understanding of the critical pathways controlling tissue repair and homeostasis, and how alloimmune responses or transplant-related processes disrupt these will lead to new immunotherapeutics that can prevent or reverse the tissue pathology leading to lost grafts due to chronic rejection.


Assuntos
Alarminas/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Transplante de Órgãos , Transdução de Sinais , Alarminas/genética , Biomarcadores , Fibrose , Regulação da Expressão Gênica , Humanos , Imunidade , Imunomodulação , Inflamação/patologia , Mediadores da Inflamação , Macrófagos/imunologia , Macrófagos/metabolismo , Transplante de Órgãos/efeitos adversos , Transplante de Órgãos/métodos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transplante Homólogo , Resultado do Tratamento
17.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-33525345

RESUMO

Chronic inflammation contributes to the development and progression of various tumors. Especially where the inflammation is mediated by cells of the innate immune system, the NLRP3 inflammasome plays an important role, as it senses and responds to a variety of exogenous and endogenous pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The NLRP3 inflammasome is responsible for the maturation and secretion of the proinflammatory cytokines interleukin-1ß (IL-1ß) and IL-18 and for the induction of a type of inflammatory cell death known as pyroptosis. Overactivation of the NLRP3 inflammasome can be a driver of various diseases. Since leukemia is known to be an inflammation-driven cancer and IL-1ß is produced in elevated levels by leukemic cells, research on NLRP3 in the context of leukemia has increased in recent years. In this review, we summarize the current knowledge on leukemia-promoting inflammation and, in particular, the role of the NLRP3 inflammasome in different types of leukemia. Furthermore, we examine a connection between NLRP3, autophagy and leukemia.


Assuntos
Carcinogênese/imunologia , Inflamassomos/imunologia , Inflamação/imunologia , Leucemia/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Alarminas/genética , Alarminas/imunologia , Animais , Autofagia/genética , Autofagia/imunologia , Carcinogênese/genética , Carcinogênese/patologia , Progressão da Doença , Regulação Leucêmica da Expressão Gênica , Humanos , Imunidade Inata , Inflamassomos/genética , Inflamação/complicações , Inflamação/genética , Inflamação/patologia , Interleucina-18/genética , Interleucina-18/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Leucemia/etiologia , Leucemia/genética , Leucemia/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Piroptose/genética , Piroptose/imunologia , Transdução de Sinais
18.
Nat Immunol ; 22(2): 154-165, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33398185

RESUMO

Inflammatory caspase sensing of cytosolic lipopolysaccharide (LPS) triggers pyroptosis and the concurrent release of damage-associated molecular patterns (DAMPs). Collectively, DAMPs are key determinants that shape the aftermath of inflammatory cell death. However, the identity and function of the individual DAMPs released are poorly defined. Our proteomics study revealed that cytosolic LPS sensing triggered the release of galectin-1, a ß-galactoside-binding lectin. Galectin-1 release is a common feature of inflammatory cell death, including necroptosis. In vivo studies using galectin-1-deficient mice, recombinant galectin-1 and galectin-1-neutralizing antibody showed that galectin-1 promotes inflammation and plays a detrimental role in LPS-induced lethality. Mechanistically, galectin-1 inhibition of CD45 (Ptprc) underlies its unfavorable role in endotoxin shock. Finally, we found increased galectin-1 in sera from human patients with sepsis. Overall, we uncovered galectin-1 as a bona fide DAMP released as a consequence of cytosolic LPS sensing, identifying a new outcome of inflammatory cell death.


Assuntos
Alarminas/metabolismo , Endotoxemia/imunologia , Galectina 1/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Alarminas/deficiência , Alarminas/genética , Animais , Estudos de Casos e Controles , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Endotoxemia/metabolismo , Endotoxemia/patologia , Feminino , Galectina 1/sangue , Galectina 1/deficiência , Galectina 1/genética , Células HeLa , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Antígenos Comuns de Leucócito/metabolismo , Lipopolissacarídeos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Necroptose , Proteínas de Ligação a Fosfato/deficiência , Proteínas de Ligação a Fosfato/genética , Células RAW 264.7 , Sepse/sangue , Sepse/diagnóstico , Transdução de Sinais , Regulação para Cima
20.
Front Immunol ; 12: 819405, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35069604

RESUMO

Breast cancer (BC) is the most common malignancy among females. Chemotherapy drugs remain the cornerstone of treatment of BC and undergo significant shifts over the past 100 years. The advent of immunotherapy presents promising opportunities and constitutes a significant complementary to existing therapeutic strategies for BC. Chemotherapy as a cytotoxic treatment that targets proliferation malignant cells has recently been shown as an effective immune-stimulus in multiple ways. Chemotherapeutic drugs can cause the release of damage-associated molecular patterns (DAMPs) from dying tumor cells, which result in long-lasting antitumor immunity by the key process of immunogenic cell death (ICD). Furthermore, Off-target effects of chemotherapy on immune cell subsets mainly involve activation of immune effector cells including natural killer (NK) cells, dendritic cells (DCs), and cytotoxic T cells, and depletion of immunosuppressive cells including Treg cells, M2 macrophages and myeloid-derived suppressor cells (MDSCs). Current mini-review summarized recent large clinical trials regarding the combination of chemotherapy and immunotherapy in BC and addressed the molecular mechanisms of immunostimulatory properties of chemotherapy in BC. The purpose of our work was to explore the immune-stimulating effects of chemotherapy at the molecular level based on the evidence from clinical trials, which might be a rationale for combinations of chemotherapy and immunotherapy in BC.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Imunidade/imunologia , Imunomodulação/efeitos dos fármacos , Imunidade Adaptativa/efeitos dos fármacos , Alarminas/genética , Alarminas/metabolismo , Animais , Antígenos de Neoplasias/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais , Ensaios Clínicos como Assunto , Gerenciamento Clínico , Suscetibilidade a Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Checkpoint Imunológico/genética , Proteínas de Checkpoint Imunológico/metabolismo , Imunidade Inata/efeitos dos fármacos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...