Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.549
Filtrar
1.
Sleep Med Clin ; 19(2): 219-228, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38692747

RESUMEN

Obstructive sleep apnea is a prevalent sleep disorder characterized by recurrent episodes of partial or complete upper airway collapse during sleep, leading to disrupted breathing patterns and intermittent hypoxia. OSA results in systemic inflammation but also directly affects the upper and lower airways leading to upregulation of inflammatory pathways and alterations of the local microbiome. These changes result in increased susceptibility to respiratory infections such as influenza, COVID-19, and bacterial pneumonia. This relationship is more complex and bidirectional in individuals with chronic lung disease such as chronic obstructive lung disease, interstitial lung disease and bronchiectasis.


Asunto(s)
Infecciones del Sistema Respiratorio , Apnea Obstructiva del Sueño , Humanos , Apnea Obstructiva del Sueño/inmunología , Apnea Obstructiva del Sueño/fisiopatología , Apnea Obstructiva del Sueño/complicaciones , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/complicaciones , Susceptibilidad a Enfermedades/inmunología , COVID-19/inmunología , COVID-19/complicaciones
2.
Viruses ; 14(10)2022 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-36298850

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with vascular endothelial cell tumor, Kaposi's sarcoma (KS) and lymphoproliferative disorder, multicentric Castleman's disease (MCD), primary effusion lymphoma (PEL) and KSHV inflammatory cytokine syndrome (KICS). Dysregulation of proinflammatory cytokines is found in most KSHV associated diseases. However, little is known about the role of host microenvironment in the regulation of KSHV establishment in B cells. In the present study, we demonstrated that IFN-γ has a strong inhibitory effect on KSHV infection but only in a subset of tonsil-derived lymphocyte samples that are intrinsically more susceptible to infection, contain higher proportions of naïve B cells, and display increased levels of IRF1 and STAT1-pY701. The effect of IFN-γ in responsive samples was associated with increased frequencies of germinal center B cells (GCB) and decreased infection of plasma cells, suggesting that IFN-γ-mediated modulation of viral dynamics in GC can inhibit the establishment of KSHV infection.


Asunto(s)
Linfocitos B , Infecciones por Herpesviridae , Herpesvirus Humano 8 , Interferón gamma , Humanos , Síndrome de Inmunodeficiencia Adquirida/inmunología , Linfocitos B/inmunología , Enfermedad de Castleman/inmunología , Citocinas/inmunología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón gamma/inmunología , Sarcoma de Kaposi/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Susceptibilidad a Enfermedades/inmunología
3.
Int J Mol Sci ; 23(6)2022 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-35328649

RESUMEN

As the global SARS-CoV-2 pandemic continues to plague healthcare systems, it has become clear that opportunistic pathogens cause a considerable proportion of SARS-CoV-2-associated mortality and morbidity cases. Of these, Covid-Associated Pulmonary Aspergilliosis (CAPA) is a major concern with evidence that it occurs in the absence of traditional risk factors such as neutropenia and is diagnostically challenging for the attending physician. In this review, we focus on the immunopathology of SARS-CoV-2 and how this potentiates CAPA through dysregulation of local and systemic immunity as well as the unintended consequences of approved COVID treatments including corticosteroids and IL-6 inhibitors. Finally, we will consider how knowledge of the above may aid in the diagnosis of CAPA using current diagnostics and what treatment should be instituted in probable and confirmed cases.


Asunto(s)
COVID-19/complicaciones , COVID-19/inmunología , Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno/inmunología , Aspergilosis Pulmonar/etiología , SARS-CoV-2/inmunología , Antifúngicos/uso terapéutico , Biomarcadores , COVID-19/virología , Manejo de la Enfermedad , Humanos , Huésped Inmunocomprometido , Aspergilosis Pulmonar/diagnóstico , Aspergilosis Pulmonar/terapia , Reproducibilidad de los Resultados , Pruebas Serológicas/métodos , Pruebas Serológicas/normas , Resultado del Tratamiento
4.
Molecules ; 27(4)2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35209238

RESUMEN

For the identification of antigenic protein biomarkers for rheumatoid arthritis (RA), we conducted IgG profiling on high density protein microarrays. Plasma IgG of 96 human samples (healthy controls, osteoarthritis, seropositive and seronegative RA, n = 24 each) and time-series plasma of a pristane-induced arthritis (PIA) rat model (n = 24 total) were probed on AIT's 16k protein microarray. To investigate the analogy of underlying disease pathways, differential reactivity analysis was conducted. A total of n = 602 differentially reactive antigens (DIRAGs) at a significance cutoff of p < 0.05 were identified between seropositive and seronegative RA for the human samples. Correlation with the clinical disease activity index revealed an inverse correlation of antibodies against self-proteins found in pathways relevant for antigen presentation and immune regulation. The PIA model showed n = 1291 significant DIRAGs within acute disease. Significant DIRAGs for (I) seropositive, (II) seronegative and (III) PIA were subjected to the Reactome pathway browser which also revealed pathways relevant for antigen presentation and immune regulation; of these, seven overlapping pathways had high significance. We therefore conclude that the PIA model reflects the biological similarities of the disease pathogenesis. Our data show that protein array analysis can elucidate biological differences and pathways relevant in disease as well be a useful additional layer of omics information.


Asunto(s)
Artritis Reumatoide/diagnóstico , Artritis Reumatoide/etiología , Autoanticuerpos/inmunología , Autoinmunidad , Biomarcadores , Animales , Autoanticuerpos/sangre , Autoantígenos/inmunología , Biología Computacional/métodos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Análisis por Matrices de Proteínas , Ratas , Índice de Severidad de la Enfermedad
5.
Mol Med ; 28(1): 18, 2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-35123388

RESUMEN

BACKGROUND: Although type 1 diabetes (T1D) is typically described as a T cell-mediated autoimmune disease, increasing evidence for a role of B cells has emerged. However, the pivotal disease-relevant B cell subset and its contribution to islet autoimmunity remain elusive. METHODS: The frequencies and phenotypic characteristics of circulating B cell subsets were analyzed using flow cytometry in individuals with new-onset T1D, long-term T1D, type 2 diabetes, and nondiabetic controls, and also in a prospective cohort of patients receiving mesenchymal stromal cell (MSC) transplantation. NOD mice and adoptive transfer assay were used to dissect the role of the certain B cell subset in disease progression. An in-vitro coculture system of islets with immune cells was established to examine the response against islets and the underlying mechanisms. RESULTS: We identified that plasmablasts, a B cell subset at the antibody-secreting stage, were significantly increased and correlated with the deterioration of beta cell function in patients with new-onset T1D. Further, a fall of plasmablast number was associated with the preservation of beta cell function in patients who received MSC transplantation after 3 months of follow-up. Meanwhile, a gradual increase of plasmablasts in pancreatic lymph nodes during the natural progression of insulitis was observed in non-obese diabetic (NOD) mice; adoptive transfer of plasmablasts together with T cells from NOD mice accelerated diabetes onset in NOD/SCID recipients. CONCLUSIONS: Our study revealed that plasmablasts may function as antigen-presenting cells and promote the activation and proinflammatory response of CD4+ T cells, further contributing to the T cell-mediated beta cell destruction. Our results provide insights into the pathogenic role of plasmablasts in islet autoimmunity and may offer new translational strategies for inhibiting T1D development.


Asunto(s)
Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Biomarcadores , Estudios de Casos y Controles , Diabetes Mellitus Tipo 1/diagnóstico , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Femenino , Humanos , Inmunohistoquímica , Inmunofenotipificación , Células Secretoras de Insulina/patología , Activación de Linfocitos , Masculino , Ratones SCID
6.
J Immunol ; 208(4): 785-792, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35115374

RESUMEN

Unlike the adaptive immune system, the innate immune system has classically been characterized as being devoid of memory functions. However, recent research shows that innate myeloid and lymphoid cells have the ability to retain memory of prior pathogen exposure and become primed to elicit a robust, broad-spectrum response to subsequent infection. This phenomenon has been termed innate immune memory or trained immunity. Innate immune memory is induced via activation of pattern recognition receptors and the actions of cytokines on hematopoietic progenitors and stem cells in bone marrow and innate leukocytes in the periphery. The trained phenotype is induced and sustained via epigenetic modifications that reprogram transcriptional patterns and metabolism. These modifications augment antimicrobial functions, such as leukocyte expansion, chemotaxis, phagocytosis, and microbial killing, to facilitate an augmented host response to infection. Alternatively, innate immune memory may contribute to the pathogenesis of chronic diseases, such as atherosclerosis and Alzheimer's disease.


Asunto(s)
Enfermedades Transmisibles/etiología , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Memoria Inmunológica , Animales , Biomarcadores , Enfermedades Transmisibles/metabolismo , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Susceptibilidad a Enfermedades/inmunología , Metabolismo Energético , Epigénesis Genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal
7.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35054960

RESUMEN

Alcoholic liver disease (ALD) is characterized by the injury, inflammation, and scarring in the liver owing to excessive alcohol consumption. Currently, ALD is a leading cause for liver transplantation. Therefore, extensive studies (in vitro, in experimental ALD models and in humans) are needed to elucidate pathological features and pathogenic mechanisms underlying ALD. Notably, oxidative changes in the liver have been recognized as a signature trait of ALD. Progression of ALD is linked to the generation of highly reactive free radicals by reactions involving ethanol and its metabolites. Furthermore, hepatic oxidative stress promotes tissue injury and, in turn, stimulates inflammatory responses in the liver, forming a pathological loop that promotes the progression of ALD. Accordingly, accumulating further knowledge on the relationship between oxidative stress and inflammation may help establish a viable therapeutic approach for treating ALD.


Asunto(s)
Biomarcadores , Susceptibilidad a Enfermedades , Hígado Graso Alcohólico/etiología , Hígado Graso Alcohólico/metabolismo , Hepatitis/complicaciones , Hepatitis/metabolismo , Estrés Oxidativo , Transducción de Señal , Animales , Susceptibilidad a Enfermedades/inmunología , Etanol/efectos adversos , Etanol/metabolismo , Hígado Graso/complicaciones , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso Alcohólico/patología , Regulación de la Expresión Génica , Hepatitis/etiología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Inmunidad Innata , Redes y Vías Metabólicas , MicroARNs/genética , Oxidación-Reducción
8.
Int J Mol Sci ; 23(2)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35054989

RESUMEN

Phenotyping cardiovascular illness and recognising heterogeneities within are pivotal in the contemporary era. Besides traditional risk factors, accumulated evidence suggested that a high inflammatory burden has emerged as a key characteristic modulating both the pathogenesis and progression of cardiovascular diseases, inclusive of atherosclerosis and myocardial infarction. To mechanistically elucidate the correlation, signalling pathways downstream to Toll-like receptors, nucleotide oligomerisation domain-like receptors, interleukins, tumour necrosis factor, and corresponding cytokines were raised as central mechanisms exerting the effect of inflammation. Other remarkable adjuvant factors include oxidative stress and secondary ferroptosis. These molecular discoveries have propelled pharmaceutical advancements. Statin was suggested to confer cardiovascular benefits not only by lowering cholesterol levels but also by attenuating inflammation. Colchicine was repurposed as an immunomodulator co-administered with coronary intervention. Novel interleukin-1ß and -6 antagonists exhibited promising cardiac benefits in the recent trials as well. Moreover, manipulation of gut microbiota and associated metabolites was addressed to antagonise inflammation-related cardiovascular pathophysiology. The gut-cardio-renal axis was therein established to explain the mutual interrelationship. As for future perspectives, artificial intelligence in conjunction with machine learning could better elucidate the sequencing of the microbiome and data mining. Comprehensively understanding the interplay between the gut microbiome and its cardiovascular impact will help identify future therapeutic targets, affording holistic care for patients with cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/terapia , Susceptibilidad a Enfermedades , Inmunomodulación , Inmunoterapia , Inflamación/complicaciones , Animales , Biomarcadores , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Retroalimentación Fisiológica , Microbioma Gastrointestinal/inmunología , Humanos , Inmunomodulación/efectos de los fármacos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Enfermedades Renales/complicaciones , Enfermedades Renales/etiología , Terapia Molecular Dirigida , Factores de Riesgo , Resultado del Tratamiento
9.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055001

RESUMEN

Non-coding RNAs, particularly lncRNAs and miRNAs, have recently been shown to regulate different steps in viral infections and induction of immune responses against viruses. Expressions of several host and viral lncRNAs have been found to be altered during viral infection. These lncRNAs can exert antiviral function via inhibition of viral infection or stimulation of antiviral immune response. Some other lncRNAs can promote viral replication or suppress antiviral responses. The current review summarizes the interaction between ncRNAs and herpes simplex virus, cytomegalovirus, and Epstein-Barr infections. The data presented in this review helps identify viral-related regulators and proposes novel strategies for the prevention and treatment of viral infection.


Asunto(s)
Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno/genética , ARN no Traducido , Virosis/etiología , Replicación Viral , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Virus ADN/fisiología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Técnicas de Diagnóstico Molecular , Unión Proteica , Especificidad de la Especie , Virosis/diagnóstico , Virosis/tratamiento farmacológico , Virosis/metabolismo , Replicación Viral/efectos de los fármacos
10.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055041

RESUMEN

Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.


Asunto(s)
Interacciones Huésped-Patógeno , Sepsis Neonatal/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/fisiología , Factores de Edad , Toxinas Bacterianas/genética , Biopelículas , Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Recién Nacido , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/prevención & control , Sepsis Neonatal/terapia , Factores de Riesgo , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/terapia , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
11.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055087

RESUMEN

Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease associated with synovial tissue proliferation, pannus formation, cartilage destruction, and systemic complications. Currently, advanced understandings of the pathologic mechanisms of autoreactive CD4+ T cells, B cells, macrophages, inflammatory cytokines, chemokines, and autoantibodies that cause RA have been achieved, despite the fact that much remains to be elucidated. This review provides an updated pathogenesis of RA which will unveil novel therapeutic targets.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Susceptibilidad a Enfermedades/inmunología , Animales , Artritis Reumatoide/diagnóstico , Artritis Reumatoide/terapia , Biomarcadores , Estudios Clínicos como Asunto , Terapia Combinada , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Técnicas de Diagnóstico Molecular , Pronóstico , Resultado del Tratamiento
12.
Mol Med ; 28(1): 10, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093033

RESUMEN

BACKGROUND: Biofilm is a community of bacteria embedded in an extracellular matrix, which can colonize different human cells and tissues and subvert the host immune reactions by preventing immune detection and polarizing the immune reactions towards an anti-inflammatory state, promoting the persistence of biofilm-embedded bacteria in the host. MAIN BODY OF THE MANUSCRIPT: It is now well established that the function of immune cells is ultimately mediated by cellular metabolism. The immune cells are stimulated to regulate their immune functions upon sensing danger signals. Recent studies have determined that immune cells often display distinct metabolic alterations that impair their immune responses when triggered. Such metabolic reprogramming and its physiological implications are well established in cancer situations. In bacterial infections, immuno-metabolic evaluations have primarily focused on macrophages and neutrophils in the planktonic growth mode. CONCLUSION: Based on differences in inflammatory reactions of macrophages and neutrophils in planktonic- versus biofilm-associated bacterial infections, studies must also consider the metabolic functions of immune cells against biofilm infections. The profound characterization of the metabolic and immune cell reactions could offer exciting novel targets for antibiofilm therapy.


Asunto(s)
Biopelículas , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/metabolismo , Interacciones Huésped-Patógeno , Infecciones/etiología , Infecciones/metabolismo , Neoplasias/complicaciones , Animales , Biopelículas/crecimiento & desarrollo , Biomarcadores , Manejo de la Enfermedad , Metabolismo Energético , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Infecciones/diagnóstico , Infecciones/terapia , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/terapia , Especificidad de Órganos
13.
Mol Cancer ; 21(1): 15, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35031075

RESUMEN

Tumor-derived exosomes (TDEs) play pivotal roles in several aspects of cancer biology. It is now evident that TDEs also favor tumor growth by negatively affecting anti-tumor immunity. As important sentinels of immune surveillance system, natural killer (NK) cells can recognize malignant cells very early and counteract the tumor development and metastasis without a need for additional activation. Based on this rationale, adoptive transfer of ex vivo expanded NK cells/NK cell lines, such as NK-92 cells, has attracted great attention and is widely studied as a promising immunotherapy for cancer treatment. However, by exploiting various strategies, including secretion of exosomes, cancer cells are able to subvert NK cell responses. This paper reviews the roles of TDEs in cancer-induced NK cells impairments with mechanistic insights. The clinical significance and potential approaches to nullify the effects of TDEs on NK cells in cancer immunotherapy are also discussed.


Asunto(s)
Exosomas/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Transporte Biológico , Biomarcadores , Comunicación Celular , Supervivencia Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Inmunoterapia , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/patología , Neoplasias/terapia , Unión Proteica , Transducción de Señal
14.
Int J Mol Sci ; 23(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35008950

RESUMEN

Mycobacterium tuberculosis (M.tb) is a successful pathogen that can reside within the alveolar macrophages of the host and can survive in a latent stage. The pathogen has evolved and developed multiple strategies to resist the host immune responses. M.tb escapes from host macrophage through evasion or subversion of immune effector functions. M.tb genome codes for PE/PPE/PE_PGRS proteins, which are intrinsically disordered, redundant and antigenic in nature. These proteins perform multiple functions that intensify the virulence competence of M.tb majorly by modulating immune responses, thereby affecting immune mediated clearance of the pathogen. The highly repetitive, redundant and antigenic nature of PE/PPE/PE_PGRS proteins provide a critical edge over other M.tb proteins in terms of imparting a higher level of virulence and also as a decoy molecule that masks the effect of effector molecules, thereby modulating immuno-surveillance. An understanding of how these proteins subvert the host immunological machinery may add to the current knowledge about M.tb virulence and pathogenesis. This can help in redirecting our strategies for tackling M.tb infections.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteínas de la Membrana/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/microbiología , Antígenos Bacterianos/química , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Susceptibilidad a Enfermedades/inmunología , Glicina/metabolismo , Humanos , Evasión Inmune , Inmunomodulación , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Mycobacterium tuberculosis/metabolismo , Virulencia
15.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35074921

RESUMEN

Proinflammatory cytokine production by innate immune cells plays a crucial role in inflammatory diseases, but the molecular mechanisms controlling the inflammatory responses are poorly understood. Here, we show that TANK-binding kinase 1 (TBK1) serves as a vital regulator of proinflammatory macrophage function and protects against tissue inflammation. Myeloid cell-conditional Tbk1 knockout (MKO) mice spontaneously developed adipose hypertrophy and metabolic disorders at old ages, associated with increased adipose tissue M1 macrophage infiltration and proinflammatory cytokine expression. When fed with a high-fat diet, the Tbk1-MKO mice also displayed exacerbated hepatic inflammation and insulin resistance, developing symptoms of nonalcoholic steatohepatitis. Furthermore, myeloid cell-specific TBK1 ablation exacerbates inflammation in experimental colitis. Mechanistically, TBK1 functions in macrophages to suppress the NF-κB and MAP kinase signaling pathways and thus attenuate induction of proinflammatory cytokines, particularly IL-1ß. Ablation of IL-1 receptor 1 (IL-1R1) eliminates the inflammatory symptoms of Tbk1-MKO mice. These results establish TBK1 as a pivotal anti-inflammatory mediator that restricts inflammation in different disease models.


Asunto(s)
Inflamación/etiología , Inflamación/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Biomarcadores , Colitis/etiología , Colitis/metabolismo , Colitis/patología , Citocinas/genética , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Regulación de la Expresión Génica , Glucosa/metabolismo , Hipertrofia , Inmunomodulación/genética , Inflamación/patología , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Interleucina-1/deficiencia , Transducción de Señal
16.
PLoS Pathog ; 18(1): e1009948, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34982799

RESUMEN

The penis is the primary site of HIV acquisition in heterosexual men. Elevated penile inflammatory cytokines increase sexual acquisition risk, and topically applied cytokines enhance foreskin HIV susceptibility in an explant model. However, the impact of penile-vaginal sex on these immune parameters is undefined. Heterosexual couples were recruited to the Sex, Couples and Science (SECS) Study, with the collection of penile swabs, semen, cervico-vaginal secretions, and blood after a period of abstinence, and repeated sampling up to 72 hours after either condomless (n = 30) or condom-protected (n = 8) penile-vaginal sex. Soluble immune parameters were quantified by multiplex immunoassay. Co-primary immune endpoints were penile levels of IL-8 and MIG, cytokines previously linked to penile HIV acquisition. One hour after sex there were dramatic increases in penile IL-8 and MIG levels, regardless of condom use, with a gradual return to baseline by 72 hours; similar patterns were observed for other chemoattractant chemokines. Penile cytokine changes were similar in circumcised and uncircumcised men, and repeated measures ANOVA and ANCOVA models demonstrated that the degree of change after condomless sex was explained by cytokine levels in their partners' cervico-vaginal secretions. This may have important implications for the biology of penile HIV acquisition.


Asunto(s)
Coito , Condones , Susceptibilidad a Enfermedades/inmunología , Infecciones por VIH/inmunología , Pene/inmunología , Adulto , Femenino , Humanos , Masculino , Sexo Inseguro , Vagina/inmunología
17.
Neoplasia ; 24(2): 145-154, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34991061

RESUMEN

A comprehensive evaluation of the clear cell renal cell carcinoma (ccRCC) immune landscape was found using 584 RNA-sequencing datasets from The Cancer Genome Atlas (TCGA), we identified 17 key dysregulated immune-associated genes in ccRCC based on association with clinical variables and important immune pathways. Of the numerous findings from our analyses, we found that several of the 17 key dysregulated genes are heavily involved in interleukin and NF-kB signaling and that somatic copy number alteration (SCNA) hotspots may be causally associated with gene dysregulation. More importantly, we also found that key immune-associated genes and pathways are strongly upregulated in ccRCC. Our study may lend novel insights into the clinical implications of immune dysregulation in ccRCC and suggests potential immunotherapeutic targets for further evaluation.


Asunto(s)
Carcinoma de Células Renales/etiología , Susceptibilidad a Enfermedades/inmunología , Neoplasias Renales/etiología , Biomarcadores , Carcinoma de Células Renales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunidad/genética , Neoplasias Renales/patología , Transducción de Señal
18.
Br J Haematol ; 196(2): 288-303, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34355382

RESUMEN

Paroxysmal nocturnal haemoglobinuria (PNH) is characterized by complement-mediated intravascular haemolysis, severe thrombophilia and bone marrow failure. While for patients with bone marrow failure the treatment follows that of immune-mediated aplastic anaemia, that of classic, haemolytic PNH is based on anti-complement medication. The anti-C5 monoclonal antibody eculizumab has revolutionized treatment, resulting in control of intravascular haemolysis and thromboembolic risk, with improved long-term survival. Novel strategies of complement inhibition are emerging. New anti-C5 agents reproduce the safety and efficacy of eculizumab, with improved patient convenience. Proximal complement inhibitors have been developed to address C3-mediated extra-vascular haemolysis and seem to improve haematological response.


Asunto(s)
Hemoglobinuria Paroxística/terapia , Algoritmos , Toma de Decisiones Clínicas , Terapia Combinada/efectos adversos , Terapia Combinada/métodos , Manejo de la Enfermedad , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/etiología , Humanos , Fenotipo , Pronóstico , Nivel de Atención , Resultado del Tratamiento
19.
Immunology ; 165(2): 143-157, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34716931

RESUMEN

Cryptococcus neoformans is an opportunistic fungal pathogen that can cause lethal cryptococcal meningitis in immunocompromised individuals such as those with HIV/AIDS. In addition, cryptococcal infections occasionally arise in immunocompetent individuals or those with previously undiagnosed immunodeficiencies. The course of cryptococcosis is highly variable in both patient groups, and there is rapidly growing evidence that genetic polymorphisms may have a significant impact on the trajectory of disease. Here, we review what is currently known about the nature of these polymorphisms and their impact on host response to C. neoformans infection. Thus far, polymorphisms in Fc gamma receptors, mannose-binding lectin, Dectin-2, Toll-like receptors and macrophage colony-stimulating factor have been associated with susceptibility to cryptococcal disease. Notably, however, in some cases the impact of these polymorphisms depends on the genetic background of the population; for example, the FCGR3A 158 F/V polymorphism was associated with an increased risk of cryptococcal disease in both HIV-positive and HIV-negative white populations, but not in Han Chinese patients. In most cases, the precise mechanism by which the identified polymorphisms influence disease progression remains unclear, although impaired fungal recognition and phagocytosis by innate immune cells appears to play a major role. Finally, we highlight outstanding questions in the field and emphasize the need for future research to include more diverse populations in their genetic association studies.


Asunto(s)
Criptococosis/etiología , Cryptococcus neoformans/inmunología , Susceptibilidad a Enfermedades/inmunología , Predisposición Genética a la Enfermedad , Huésped Inmunocomprometido , Fenómenos Inmunogenéticos , Polimorfismo Genético , Inmunidad Adaptativa , Animales , Biomarcadores , Criptococosis/metabolismo , Regulación de la Expresión Génica , Variación Genética , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Transducción de Señal
20.
Immunology ; 165(1): 22-43, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34704249

RESUMEN

Neutrophil-centred inflammation and flawed clearance of neutrophils cause and exuberate multiple pathological conditions. These most abundant leukocytes exhibit very high daily turnover in steady-state and stress conditions. Various armours including oxidative burst, NETs and proteases function against pathogens, but also dispose neutrophils to spawn pro-inflammatory responses. Neutrophils undergo death through different pathways upon ageing, infection, executing the intruder's elimination. These include non-lytic apoptosis and other lytic deaths including NETosis, necroptosis and pyroptosis with distinct disintegration of the cellular membrane. This causes release and presence of different intracellular cytotoxic, and tissue-damaging content as cell remnants in the extracellular environment. The apoptotic cells and apoptotic bodies get cleared with non-inflammatory outcomes, while lytic deaths associated remnants including histones and cell-free DNA cause pro-inflammatory responses. Indeed, the enhanced frequencies of neutrophil-associated proteases, cell-free DNA and autoantibodies in diverse pathologies including sepsis, asthma, lupus and rheumatoid arthritis, imply disturbed neutrophil resolution programmes in inflammatory and autoimmune diseases. Thus, the clearance mechanisms of neutrophils and associated remnants are vital for therapeutics. Though studies focused on clearance mechanisms of senescent or apoptotic neutrophils so far generated a good understanding of the same, clearance of neutrophils undergoing distinct lytic deaths, including NETs, are being the subjects of intense investigations. Here, in this review, we are providing the current updates in the clearance mechanisms of apoptotic neutrophils and focusing on not so well-defined recognition, uptake and degradation of neutrophils undergoing lytic death and associated remnants that may provide new therapeutic approaches in inflammation and autoimmunity.


Asunto(s)
Inmunidad , Inflamación/etiología , Inflamación/metabolismo , Neutrófilos/fisiología , Animales , Apoptosis/inmunología , Biomarcadores , Susceptibilidad a Enfermedades/inmunología , Trampas Extracelulares/inmunología , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...