Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Curr Opin Immunol ; 90: 102457, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39232338

RESUMEN

The innate immune system employs two different strategies to detect pathogens: first, it recognizes microbial components as ligands of pattern recognition receptors (pattern-triggered immunity [PTI]), and second, it detects the activities of pathogen-encoded effectors (effector-triggered immunity [ETI]). Recently, these pathogen-centric concepts were expanded to include sensing of self-derived signals during cellular distress or damage (damage-triggered immunity [DTI]). This extension relied on broadening the PTI model to include damage-associated molecular patterns (DAMPs). However, applying the pattern recognition framework of PTI to DTI overlooks the critical role of sterile activation of ETI pathways. We argue that both PTI and ETI pathways are prone to erroneous detection of self, which is largely attributable to 'friendly fire' rather than protective immune activation. This erroneous activation is inherent to the trade-off between sensitivity and specificity of immune sensing and might be tolerated because its detrimental effects emerge late in life, a phenomenon known as antagonistic pleiotropy.


Asunto(s)
Inmunidad Innata , Receptores de Reconocimiento de Patrones , Humanos , Animales , Receptores de Reconocimiento de Patrones/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Alarminas/inmunología , Alarminas/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Transducción de Señal/inmunología , Interacciones Huésped-Patógeno/inmunología
3.
Inflamm Res ; 73(7): 1239-1252, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38844678

RESUMEN

BACKGROUND: We have previously shown that asthma-like airways inflammation may be induced by topical exposure to respiratory tract pathogens such as S. pneumoniae (SP) in concert with epithelial alarmins such as IL-33. Details of the pathogenesis of this murine surrogate remain however unexplored. METHODS: Airways inflammation was induced by repeated, intranasal exposure of Il-4-/-, Rag1-/- and Rag2-/-Il2rg-/- mice (in which B lymphocyte IgE switching, adaptive and innate immunity are respectively ablated) as well as wild type mice to inactivated SP, IL-33 or both. Airways pathological changes were analysed, and the subsets and functions of locally accumulated ILC2s investigated by single cell RNA sequencing and flow cytometry. RESULTS: In the presence of IL-33, repeated exposure of the airways to inactivated SP caused marked eosinophil- and neutrophil-rich inflammation and local accumulation of ILC2s, which was retained in the Il-4-/- and Rag1-/- deficient mice but abolished in the Rag2-/-Il2rg-/- mice, an effect partly reversed by adoptive transfer of ILC2s. Single cell sequencing analysis of ILC2s recruited following SP and IL-33 exposure revealed a Klrg1+Ly6a+subset, expressing particularly elevated quantities of the pro-inflammatory cytokine IL-6, type 2 cytokines (IL-5 and IL-13) and MHC class II molecules, promoting type 2 inflammation as well as involved in neutrophil-mediated inflammatory responses. CONCLUSION: Local accumulation of KLRG1+Ly6a+ ILC2s in the lung tissue is a critical aspect of the pathogenesis of airways eosinophilic and neutrophil-rich inflammation induced by repeated exposure to SP in the presence of the epithelial alarmin IL-33.


Asunto(s)
Interleucina-33 , Streptococcus pneumoniae , Animales , Interleucina-33/inmunología , Interleucina-33/genética , Streptococcus pneumoniae/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Pulmón/inmunología , Pulmón/patología , Pulmón/microbiología , Linfocitos/inmunología , Inflamación/inmunología , Ratones , Femenino , Alarminas/inmunología , Proteínas de Homeodominio
4.
Front Immunol ; 15: 1403018, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38881893

RESUMEN

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease primarily affecting premature neonates, marked by poorly understood pro-inflammatory signaling cascades. Recent advancements have shed light on a subset of endogenous molecular patterns, termed chromatin-associated molecular patterns (CAMPs), which belong to the broader category of damage-associated molecular patterns (DAMPs). CAMPs play a crucial role in recognizing pattern recognition receptors and orchestrating inflammatory responses. This review focuses into the realm of CAMPs, highlighting key players such as extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), cell-free DNA, neutrophil extracellular traps (NETs), histones, and extracellular RNA. These intrinsic molecules, often perceived as foreign, have the potential to trigger immune signaling pathways, thus contributing to NEC pathogenesis. In this review, we unravel the current understanding of the involvement of CAMPs in both preclinical and clinical NEC scenarios. We also focus on elucidating the downstream signaling pathways activated by these molecular patterns, providing insights into the mechanisms that drive inflammation in NEC. Moreover, we scrutinize the landscape of targeted therapeutic approaches, aiming to mitigate the impact of tissue damage in NEC. This in-depth exploration offers a comprehensive overview of the role of CAMPs in NEC, bridging the gap between preclinical and clinical insights.


Asunto(s)
Alarminas , Cromatina , Enterocolitis Necrotizante , Humanos , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/inmunología , Alarminas/metabolismo , Alarminas/inmunología , Cromatina/metabolismo , Animales , Transducción de Señal , Recién Nacido , Proteína HMGB1/metabolismo
5.
Front Immunol ; 15: 1367053, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756775

RESUMEN

Background: With the worsening of the greenhouse effect, the correlation between the damp-heat environment (DH) and the incidence of various diseases has gained increasing attention. Previous studies have demonstrated that DH can lead to intestinal disorders, enteritis, and an up-regulation of NOD-like receptor protein 3 (NLRP3). However, the mechanism of NLRP3 in this process remains unclear. Methods: We established a DH animal model to observe the impact of a high temperature and humidity environment on the mice. We sequenced the 16S rRNA of mouse feces, and the RNA transcriptome of intestinal tissue, as well as the levels of cytokines including interferon (IFN)-γ and interleukin (IL)-4 in serum. Results: Our results indicate that the intestinal macrophage infiltration and the expression of inflammatory genes were increased in mice challenged with DH for 14 days, while the M2 macrophages were decreased in Nlrp3 -/- mice. The alpha diversity of intestinal bacteria in Nlrp3 -/- mice was significantly higher than that in control mice, including an up-regulation of the Firmicutes/Bacteroidetes ratio. Transcriptomic analysis revealed 307 differentially expressed genes were decreased in Nlrp3 -/- mice compared with control mice, which was related to humoral immune response, complement activation, phagocytic recognition, malaria and inflammatory bowel disease. The ratio of IFN-γ/IL-4 was decreased in control mice but increased in Nlrp3 -/- mice. Conclusions: Our study found that the inflammation induced by DH promotes Th2-mediated immunity via NLRP3, which is closely related to the disruption of intestinal flora.


Asunto(s)
Microbioma Gastrointestinal , Calor , Proteína con Dominio Pirina 3 de la Familia NLR , Células Th2 , Animales , Ratones , Alarminas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/inmunología , Macrófagos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Células Th2/inmunología
6.
J Thromb Haemost ; 22(8): 2140-2146, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38815754

RESUMEN

It is increasingly apparent that the pathologic interplay between coagulation and innate immunity, ie, immunothrombosis, forms the common basis of many challenges across the boundaries of specialized medicine and cannot be fully explained by the conventional concepts of cascade and cell-based coagulation. To improve our understanding of coagulation, we propose a model of coagulation that converges with inflammation and innate immune activation as a unified response toward vascular injury. Evolutionarily integral to the convergent response are damage-associated molecular patterns, which are released as a consequence of injury. Damage-associated molecular patterns facilitate diverse interactions within and between systems, not only to complement and reinforce cell-based clot formation but also to steer the response toward clot resolution and wound healing. By extending coagulation beyond its current boundaries, the convergent model aims to deliver novel diagnostics and therapeutics for contemporary and unexpected challenges across medicine, as exposed by COVID-19 and vaccine-induced immune thrombotic thrombocytopenia.


Asunto(s)
Coagulación Sanguínea , COVID-19 , Inmunidad Innata , Humanos , COVID-19/inmunología , COVID-19/sangre , Trombosis/inmunología , Trombosis/sangre , Animales , Inflamación/inmunología , SARS-CoV-2/inmunología , Plaquetas/inmunología , Plaquetas/metabolismo , Alarminas/metabolismo , Alarminas/inmunología
7.
Nat Rev Immunol ; 24(10): 703-719, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38684933

RESUMEN

Damage-associated molecular patterns (DAMPs) are endogenous molecules that are released from host cells as a result of cell death or damage. The release of DAMPs in tissues is associated with loss of tissue homeostasis. Sensing of DAMPs by innate immune receptors triggers inflammation, which can be beneficial in initiating the processes that restore tissue homeostasis but can also drive inflammatory diseases. In recent years, the sensing of intracellular DAMPs has received extensive attention in the field of sterile inflammation. However, emerging studies have shown that DAMPs that originate from neighbouring cells, and even from distal tissues or organs, also mediate sterile inflammatory responses. This multi-level sensing of DAMPs is crucial for intercellular, trans-tissue and trans-organ communication. Here, we summarize how DAMP-sensing receptors detect DAMPs from intracellular, intercellular or distal tissue and organ sources to mediate sterile inflammation. We also discuss the possibility of targeting DAMPs or their corresponding receptors to treat inflammatory diseases.


Asunto(s)
Alarminas , Inflamación , Transducción de Señal , Humanos , Inflamación/inmunología , Alarminas/inmunología , Alarminas/metabolismo , Animales , Transducción de Señal/inmunología , Inmunidad Innata/inmunología
9.
JCI Insight ; 9(8)2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470489

RESUMEN

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.


Asunto(s)
Linfocitos B , Animales , Ratones , Linfocitos B/inmunología , Células Th2/inmunología , Modelos Animales de Enfermedad , Ratones Transgénicos , Ratones Noqueados , Inmunidad Innata/inmunología , Ratones Endogámicos C57BL , Inmunoglobulina E/inmunología , Alarminas/inmunología , Anticuerpos/inmunología , Hipersensibilidad/inmunología , Eosinofilia/inmunología
10.
Immunity ; 55(4): 623-638.e5, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35385697

RESUMEN

The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.


Asunto(s)
Alarminas , Mucosa Intestinal , Acilación , Alarminas/inmunología , Antihelmínticos/inmunología , Biomarcadores de Tumor , Citocinas , Proteínas de Unión al ADN , Helmintiasis/inmunología , Humanos , Hiperplasia , Inflamación , Interleucina-33 , Mucosa Intestinal/inmunología , Mebendazol , N-Acetilglucosaminiltransferasas/inmunología , Proteínas Citotóxicas Formadoras de Poros , Factor de Transcripción STAT6/inmunología
11.
J Hematol Oncol ; 15(1): 5, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35012610

RESUMEN

BACKGROUND: Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19) through direct lysis of infected lung epithelial cells, which releases damage-associated molecular patterns and induces a pro-inflammatory cytokine milieu causing systemic inflammation. Anti-viral and anti-inflammatory agents have shown limited therapeutic efficacy. Soluble CD24 (CD24Fc) blunts the broad inflammatory response induced by damage-associated molecular patterns via binding to extracellular high mobility group box 1 and heat shock proteins, as well as regulating the downstream Siglec10-Src homology 2 domain-containing phosphatase 1 pathway. A recent randomized phase III trial evaluating CD24Fc for patients with severe COVID-19 (SAC-COVID; NCT04317040) demonstrated encouraging clinical efficacy. METHODS: Using a systems analytical approach, we studied peripheral blood samples obtained from patients enrolled at a single institution in the SAC-COVID trial to discern the impact of CD24Fc treatment on immune homeostasis. We performed high dimensional spectral flow cytometry and measured the levels of a broad array of cytokines and chemokines to discern the impact of CD24Fc treatment on immune homeostasis in patients with COVID-19. RESULTS: Twenty-two patients were enrolled, and the clinical characteristics from the CD24Fc vs. placebo groups were matched. Using high-content spectral flow cytometry and network-level analysis, we found that patients with severe COVID-19 had systemic hyper-activation of multiple cellular compartments, including CD8+ T cells, CD4+ T cells, and CD56+ natural killer cells. Treatment with CD24Fc blunted this systemic inflammation, inducing a return to homeostasis in NK and T cells without compromising the anti-Spike protein antibody response. CD24Fc significantly attenuated the systemic cytokine response and diminished the cytokine coexpression and network connectivity linked with COVID-19 severity and pathogenesis. CONCLUSIONS: Our data demonstrate that CD24Fc rapidly down-modulates systemic inflammation and restores immune homeostasis in SARS-CoV-2-infected individuals, supporting further development of CD24Fc as a novel therapeutic against severe COVID-19.


Asunto(s)
Antígeno CD24/uso terapéutico , COVID-19/prevención & control , Síndrome de Liberación de Citoquinas/prevención & control , Inflamación/prevención & control , SARS-CoV-2/efectos de los fármacos , Anciano , Alarminas/inmunología , Alarminas/metabolismo , Antígeno CD24/química , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Método Doble Ciego , Femenino , Proteína HMGB1/inmunología , Proteína HMGB1/metabolismo , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/virología , Masculino , Persona de Mediana Edad , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Solubilidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología , Resultado del Tratamiento
12.
Cell Immunol ; 371: 104470, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942481

RESUMEN

Inflammatory responses are required to block pathogen infection but can also lead to hypersensitivity and chronic inflammation. Barrier tissues actively release IL-33, ATP, and other alarmins during cell stress, helping identify pathogenic stimuli. However, it is unclear how these signals are integrated. Mast cells are critical initiators of allergic inflammation and respond to IL-33 and ATP. We found that mouse mast cells had a 3-6-fold increase in ATP-induced cytokine production when pre-treated with IL-33. This effect was observed at ATP concentrations < 100 µM and required < 30-minute IL-33 exposure. ATP-induced degranulation was not enhanced by pretreatment nor was the response to several pathogen molecules. Mechanistic studies implicated the P2X7 receptor and calcineurin/NFAT pathway in the enhanced ATP response. Finally, we found that IL-33 + ATP co-stimulation enhanced peritoneal eosinophil and macrophage recruitment. These results support the hypothesis that alarmins collaborate to surpass a threshold necessary to initiate an inflammatory response.


Asunto(s)
Adenosina Trifosfato/metabolismo , Alarminas/inmunología , Interleucina-33/metabolismo , Mastocitos/metabolismo , Peritonitis/patología , Animales , Calcineurina/metabolismo , Degranulación de la Célula/inmunología , Células Cultivadas , Citocinas/biosíntesis , Eosinófilos/inmunología , Inflamación/patología , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo
13.
Front Immunol ; 12: 783616, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899749

RESUMEN

Idiopathic inflammatory myopathy (IIM) is a heterogeneous group of acquired, autoimmune muscle diseases characterized by muscle inflammation and extramuscular involvements. Present literatures have revealed that dysregulated cell death in combination with impaired elimination of dead cells contribute to the release of autoantigens, damage-associated molecular patterns (DAMPs) and inflammatory cytokines, and result in immune responses and tissue damages in autoimmune diseases, including IIMs. This review summarizes the roles of various forms of programmed cell death pathways in the pathogenesis of IIMs and provides evidence for potential therapeutic targets.


Asunto(s)
Músculo Esquelético/patología , Miositis/patología , Muerte Celular Regulada , Alarminas/inmunología , Alarminas/metabolismo , Animales , Autoantígenos/inmunología , Autoantígenos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Miositis/inmunología , Miositis/metabolismo , Transducción de Señal
14.
Front Immunol ; 12: 740548, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721407

RESUMEN

Certain cancer therapy has been shown to induce immunogenic cell death in cancer cells and may promote tumor progression instead. The external stress or stimuli may induce cell death and contribute toward the secretion of pro inflammatory molecules. The release of damage-associated molecular patterns (DAMPs) upon induction of therapy or cell death has been shown to induce an inflammatory response. Nevertheless, the mechanism as to how the DAMPs are released and engage in such activity needs further in-depth investigation. Interestingly, some studies have shown that DAMPs can be released through extracellular vesicles (EVs) and can bind to receptors such as toll-like receptors (TCRs). Ample pre-clinical studies have shown that cancer-derived EVs are able to modulate immune responses within the tumor microenvironment. However, the information on the presence of such DAMPs within EVs is still elusive. Therefore, this mini-review attempts to summarize and appraise studies that have shown the presence of DAMPs within cancer-EVs and how it affects the downstream cellular process.


Asunto(s)
Alarminas/inmunología , Vesículas Extracelulares/metabolismo , Neoplasias/inmunología , Animales , Carcinogénesis , Humanos , Inmunidad , Inmunomodulación , Neoplasias/terapia , Receptores Toll-Like/metabolismo , Microambiente Tumoral
15.
J Neuroinflammation ; 18(1): 252, 2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34727934

RESUMEN

OBJECTIVE: Astrocytes participate in the local innate immune response of the central nervous system. In response to stress such as ischemia, activated cells release endogenous factors known as damage-associated molecular patterns (DAMPs). Self-extracellular RNA (eRNA) is such a ubiquitous alarm signal. However, it is unclear whether eRNA is involved in the early acute phase of cerebral ischemia and is sufficient to sensitize astrocytes towards a DAMP or PAMP (pathogen-associated molecular pattern) reaction. METHODS: Pro-inflammatory activation upon eRNA stimulation was characterized in primary murine astrocyte cultures. In vivo, an experimental stroke model was used to localize and quantify eRNA in murine brain sections. Using primary cortical neurons and the mouse hippocampal neuronal cell line HT-22, neuronal RNA release upon stress conditions related to cerebral hypoxia/ischemia was analyzed. RESULTS: While low-dose eRNA alone did not promote pro-inflammatory activation of astrocytes in culture, it strongly enhanced the expression of pro-inflammatory cytokines in the presence of either Pam2CSK4, a synthetic PAMP molecule that mimics bacterial infection, or high mobility group box 1 (HMGB1), a prominent DAMP. Synergism of eRNA/Pam2CSK4 and eRNA/HMGB1 was prevented by blockage of the astroglial toll-like receptor (TLR)-2. Inhibition of NF-κB- and mitogen-activated protein kinase-dependent signaling pathways hampered eRNA/Pam2CSK4-mediated pro-inflammatory activation of astrocytes. In vivo, the amount of non-nuclear, presumably extracellular ribosomal RNA in close proximity to neurons significantly accumulated across the infarct core and peri-infarct areas that was accompanied by transcriptional up-regulation of various pro-inflammatory factors. Accordingly, the exposure of neurons to hypoxic/ischemic stress in vitro resulted in the release of eRNA, partly mediated by active cellular processes dependent on the cytosolic calcium level. CONCLUSION: The DAMP signal eRNA can sensitize astrocytes as active players in cerebral innate immunity towards exogenous and endogenous activators of inflammation (PAMPs and DAMPs) in a synergistic manner via TLR2-NF-κB-dependent signaling mechanisms. These findings provide new insights into the pathogenesis of ischemic stroke and other inflammatory neurological disorders. Further studies will clarify whether administration of RNase in vivo may serve as an effective treatment for inflammatory brain pathologies.


Asunto(s)
Alarminas/inmunología , Astrocitos/inmunología , Inflamación/inmunología , ARN/inmunología , Accidente Cerebrovascular/inmunología , Animales , Ratones , Accidente Cerebrovascular/patología
16.
Front Immunol ; 12: 754127, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777366

RESUMEN

COVID-19 presentations range from mild to moderate through severe disease but also manifest with persistent illness or viral recrudescence. We hypothesized that the spectrum of COVID-19 disease manifestations was a consequence of SARS-CoV-2-mediated delay in the pathogen-associated molecular pattern (PAMP) response, including dampened type I interferon signaling, thereby shifting the balance of the immune response to be dominated by damage-associated molecular pattern (DAMP) signaling. To test the hypothesis, we constructed a parsimonious mechanistic mathematical model. After calibration of the model for initial viral load and then by varying a few key parameters, we show that the core model generates four distinct viral load, immune response and associated disease trajectories termed "patient archetypes", whose temporal dynamics are reflected in clinical data from hospitalized COVID-19 patients. The model also accounts for responses to corticosteroid therapy and predicts that vaccine-induced neutralizing antibodies and cellular memory will be protective, including from severe COVID-19 disease. This generalizable modeling framework could be used to analyze protective and pathogenic immune responses to diverse viral infections.


Asunto(s)
Alarminas/inmunología , Tratamiento Farmacológico de COVID-19 , COVID-19 , Modelos Biológicos , SARS-CoV-2 , Corticoesteroides/uso terapéutico , Adulto , Anciano , Antiinflamatorios/uso terapéutico , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/diagnóstico , COVID-19/inmunología , COVID-19/virología , Vacunas contra la COVID-19 , Humanos , Persona de Mediana Edad , Reproducibilidad de los Resultados , Carga Viral
17.
Front Immunol ; 12: 744927, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34621275

RESUMEN

Ischemia and reperfusion injury is an early inflammatory process during liver transplantation that impacts on graft function and clinical outcomes. Interleukin (IL)-33 is a danger-associated molecular pattern involved in kidney ischemia/reperfusion injury and several liver diseases. The aims were to assess whether IL-33 was released as an alarmin responsible for ischemia/reperfusion injury in a mouse model of warm hepatic ischemia, and whether this hypothesis could also apply in the setting of human liver transplantation. First, a model of warm hepatic ischemia/reperfusion was used in wild-type and IL-33-deficient mice. Severity of ischemia/reperfusion injury was assessed with ALT and histological analysis. Then, serum IL-33 was measured in a pilot cohort of 40 liver transplant patients. Hemodynamic postreperfusion syndrome, graft dysfunction (assessed by model for early allograft scoring >6), renal failure, and tissue lesions on time-zero biopsies were assessed. In the mouse model, IL-33 was constitutively expressed in the nucleus of endothelial cells, immediately released in response to hepatic pedicle clamping without neosynthesis, and participated in the recruitment of neutrophils and tissue injury on site. The kinetics of IL-33 in liver transplant patients strikingly matched the ones in the animal model, as attested by serum levels reaching a peak immediately after reperfusion, which correlated to clinical outcomes including postreperfusion syndrome, posttransplant renal failure, graft dysfunction, and histological lesions of ischemia/reperfusion injury. IL-33 was an independent factor of graft dysfunction with a cutoff of IL-33 at 73 pg/ml after reperfusion (73% sensitivity, area under the curve of 0.76). Taken together, these findings establish the immediate implication of IL-33 acting as an alarmin in liver I/R injury and provide evidence of its close association with cardinal features of early liver injury-associated disorders in LT patients.


Asunto(s)
Alarminas/inmunología , Interleucina-33/inmunología , Trasplante de Hígado , Hígado/patología , Daño por Reperfusión/patología , Alarminas/metabolismo , Animales , Estudios de Cohortes , Humanos , Interleucina-33/metabolismo , Hígado/inmunología , Hígado/metabolismo , Ratones , Proyectos Piloto , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo
18.
Cell Immunol ; 369: 104424, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34469845

RESUMEN

Invading pathogens are contained/eliminated by orchestrated actions of different humoral components of the innate immune response. One of them is endogenous molecules called alarmins, which contribute to diverse processes from danger sense until the infection extinction. Considering the participation of mast cells (MCs) in many aspects of the body's defense and, on the other hand, the importance of alarmins as molecules that signal damage/danger, in this study, we evaluated the effect of alarmins on MC phenotype and activity. We found that cathelicidin CRAMP and cytokine IL-33 significantly affect the appearance of Dectin-1, Dectin-2, RIG-I, and NOD1 receptors in mature MCs and modulate their inflammatory response. We established that chosen alarmins might stimulate MCs to release pro-inflammatory and immunoregulatory mediators and induce a migratory response. In conclusion, our data highlight that alarmins CRAMP and IL-33 might strongly influence MC features and activity, mainly by strengthening their role in the inflammatory mechanisms and controlling the activity of cells participating in antimicrobial processes.


Asunto(s)
Alarminas/metabolismo , Catelicidinas/metabolismo , Interleucina-33/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Adaptación Fisiológica/inmunología , Alarminas/inmunología , Animales , Catelicidinas/inmunología , Movimiento Celular/inmunología , Femenino , Inmunidad Innata/inmunología , Interleucina-33/inmunología , Ratas , Ratas Wistar
19.
Front Immunol ; 12: 628822, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381442

RESUMEN

Background: Skeletal muscle ischemia/reperfusion (I/R) injury is an important clinical issue that can cause remote organ injury. Although its pathogenesis has not been fully elucidated, recent studies have suggested that damage-associated molecular patterns (DAMPs) are mediators of remote organ injury in sterile inflammation. The purpose of this study was to investigate the possible involvement of DAMPs, including the nuclear proteins high-mobility group box 1 (HMGB1) and histone H3, in the pathogenesis of skeletal muscle I/R injury in mice. Methods: Hindlimb ischemia was induced in mice through bilateral ligation of inguinal regions using rubber grommets. Reperfusion was induced by cutting the rubber grommets after 2-12 h of ischemic period. Survival rates, localization of HMGB1 and histone H3 in the gastrocnemius muscle, and circulating HMGB1 and histone H3 levels were analyzed. The effect of anti-HMGB1 and anti-histone H3 antibodies on survival was analyzed in mice with I/R injury. Results: All mice with hindlimb ischemia survived for at least 36 h, while all mice died within 24 h if the hindlimbs were reperfused after ischemia for 4-12 h. Immunohistochemical analysis revealed that HMGB1 translocated from the nucleus to the cytoplasm in the ischemic gastrocnemius muscle, while histone H3 was confined to the nucleus. Accordingly, serum HMGB1 levels were significantly elevated in mice with hindlimb I/R compared with normal mice or mice with hindlimb ischemia (P < 0.05). Serum histone H3 levels were not elevated after I/R. Treatment with anti-HMGB1 antibodies significantly improved survival of mice with hindlimb I/R injury compared with control antibodies (P < 0.05). Conclusions: HMGB1, but not histone H3, translocated to the cytoplasm during skeletal muscle ischemia, and was released into the systemic circulation after reperfusion in mice with I/R injury. Treatment with anti-HMGB1 antibodies partially improved survival.


Asunto(s)
Alarminas/metabolismo , Proteína HMGB1/metabolismo , Miembro Posterior/patología , Histonas/metabolismo , Músculo Esquelético/fisiología , Daño por Reperfusión/metabolismo , Alarminas/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Modelos Animales de Enfermedad , Proteína HMGB1/inmunología , Miembro Posterior/cirugía , Histonas/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Daño por Reperfusión/inmunología
20.
Front Immunol ; 12: 720192, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34456928

RESUMEN

COVID-19 might lead to multi-organ failure and, in some cases, to death. The COVID-19 severity is associated with a "cytokine storm." Danger-associated molecular patterns (DAMPs) are proinflammatory molecules that can activate pattern recognition receptors, such as toll-like receptors (TLRs). DAMPs and TLRs have not received much attention in COVID-19 but can explain some of the gender-, weight- and age-dependent effects. In females and males, TLRs are differentially expressed, likely contributing to higher COVID-19 severity in males. DAMPs and cytokines associated with COVID-19 mortality are elevated in obese and elderly individuals, which might explain the higher risk for severer COVID-19 in these groups. Adenosine signaling inhibits the TLR/NF-κB pathway and, through this, decreases inflammation and DAMPs' effects. As vaccines will not be effective in all susceptible individuals and as new vaccine-resistant SARS-CoV-2 mutants might develop, it remains mandatory to find means to dampen COVID-19 disease severity, especially in high-risk groups. We propose that the regulation of DAMPs via adenosine signaling enhancement might be an effective way to lower the severity of COVID-19 and prevent multiple organ failure in the absence of severe side effects.


Asunto(s)
Alarminas/inmunología , COVID-19/fisiopatología , Mediadores de Inflamación/inmunología , Adenosina/metabolismo , Alarminas/antagonistas & inhibidores , Animales , COVID-19/complicaciones , COVID-19/inmunología , COVID-19/terapia , Humanos , Inflamación/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Insuficiencia Multiorgánica/etiología , Insuficiencia Multiorgánica/prevención & control , Gravedad del Paciente , Transducción de Señal , Receptores Toll-Like/antagonistas & inhibidores , Receptores Toll-Like/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA