Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 623
Filtrar
1.
Sci Rep ; 14(1): 19353, 2024 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-39169069

RESUMEN

Infectious endophthalmitis (IE) poses a significant threat to vision. This study aimed to explore the impact of microRNA (miR)-27a-3p on inflammation in IE. A rat model was developed through intravitreal injection of lipopolysaccharide. Clinical and demographic data were collected for 54 participants: 31 diagnosed with IE and 23 non-infectious patients with idiopathic macular holes. Expression levels of miR-27a-3p and inflammatory genes were quantified via reverse transcription quantitative polymerase chain reaction. Concentrations of inflammatory cytokines in human vitreous samples were measured using enzyme-linked immunosorbent assay. In vitro studies were conducted to explore the target gene of miR-27a-3p. The final animal experiments further verified the role of miR-27a-3p and tuberous sclerosis complex (TSC)1 in inflammatory responses. Results showed that miR-27a-3p was elevated in LPS-treated rats and IE patients. Thirty-one IE patients were divided into the High (n = 15) and Low (n = 16) groups according to the expression of miR-27a-3p. No significant differences were observed in baseline clinical and demographic characteristics between the control and IE patient groups. Pro-inflammatory cytokine mRNA levels and concentrations were notably increased in both LPS-treated rats and the High group of patients. Besides, results showed that TSC1 is a target gene of miR-27a-3p. Moreover, TSC1 inhibition promoted inflammation in rat vitreous samples. In summary, our findings suggested that miR-27a-3p exacerbated inflammatory responses in IE though targeting TSC1, offering novel insights for potential therapeutic strategies targeting miR-27a-3p in the clinical management of IE.


Asunto(s)
Endoftalmitis , Inflamación , MicroARNs , Proteína 1 del Complejo de la Esclerosis Tuberosa , MicroARNs/genética , MicroARNs/metabolismo , Animales , Humanos , Endoftalmitis/metabolismo , Endoftalmitis/genética , Endoftalmitis/patología , Ratas , Masculino , Femenino , Proteína 1 del Complejo de la Esclerosis Tuberosa/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Inflamación/genética , Inflamación/metabolismo , Anciano , Lipopolisacáridos , Citocinas/metabolismo , Citocinas/genética , Persona de Mediana Edad , Modelos Animales de Enfermedad , Ratas Sprague-Dawley
2.
Adv Rheumatol ; 64(1): 62, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39175060

RESUMEN

Systemic autoinflammatory diseases (SAIDs) arise from dysregulated innate immune system activity, which leads to systemic inflammation. These disorders, encompassing a diverse array of genetic defects classified as inborn errors of immunity, are significant diagnostic challenges due to their genetic heterogeneity and varied clinical presentations. Although recent advances in genetic sequencing have facilitated pathogenic gene discovery, approximately 40% of SAIDs patients lack molecular diagnoses. SAIDs have distinct clinical phenotypes, and targeted therapeutic approaches are needed. This review aims to underscore the complexity and clinical significance of SAIDs, focusing on prototypical disorders grouped according to their pathophysiology as follows: (i) inflammasomopathies, characterized by excessive activation of inflammasomes, which induces notable IL-1ß release; (ii) relopathies, which are monogenic disorders characterized by dysregulation within the NF-κB signaling pathway; (iii) IL-18/IL-36 signaling pathway defect-induced SAIDs, autoinflammatory conditions defined by a dysregulated balance of IL-18/IL-36 cytokine signaling, leading to uncontrolled inflammation and tissue damage, mainly in the skin; (iv) type I interferonopathies, a diverse group of disorders characterized by uncontrolled production of type I interferons (IFNs), notably interferon α, ß, and ε; (v) anti-inflammatory signaling pathway impairment-induced SAIDs, a spectrum of conditions characterized by IL-10 and TGFß anti-inflammatory pathway disruption; and (vi) miscellaneous and polygenic SAIDs. The latter group includes VEXAS syndrome, chronic recurrent multifocal osteomyelitis/chronic nonbacterial osteomyelitis, Schnitzler syndrome, and Still's disease, among others, illustrating the heterogeneity of SAIDs and the difficulty in creating a comprehensive classification. Therapeutic strategies involving targeted agents, such as JAK inhibitors, IL-1 blockers, and TNF inhibitors, are tailored to the specific disease phenotypes.


Asunto(s)
Enfermedades Autoinflamatorias Hereditarias , Humanos , Enfermedades Autoinflamatorias Hereditarias/genética , Enfermedades Autoinflamatorias Hereditarias/tratamiento farmacológico , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Inflamasomas/genética , Inflamación/genética , Transducción de Señal , Interleucina-18/genética , Interleucina-1beta/genética , Interleucina-1beta/antagonistas & inhibidores , FN-kappa B , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/terapia , Anemia Diseritropoyética Congénita/diagnóstico , Síndrome de Schnitzler/genética , Síndrome de Schnitzler/tratamiento farmacológico , Síndrome de Schnitzler/diagnóstico , Osteomielitis/genética , Osteomielitis/tratamiento farmacológico , Osteomielitis/inmunología , Deficiencia de Mevalonato Quinasa/genética , Deficiencia de Mevalonato Quinasa/tratamiento farmacológico , Deficiencia de Mevalonato Quinasa/diagnóstico , Síndromes de Inmunodeficiencia
3.
Nat Cell Biol ; 26(8): 1336-1345, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39103548

RESUMEN

The accumulation of senescent cells promotes ageing and age-related diseases, but molecular mechanisms that senescent cells use to evade immune clearance and accumulate in tissues remain to be elucidated. Here we report that p16-positive senescent cells upregulate the immune checkpoint protein programmed death-ligand 1 (PD-L1) to accumulate in ageing and chronic inflammation. We show that p16-mediated inhibition of cell cycle kinases CDK4/6 induces PD-L1 stability in senescent cells via downregulation of its ubiquitin-dependent degradation. p16-expressing senescent alveolar macrophages elevate PD-L1 to promote an immunosuppressive environment that can contribute to an increased burden of senescent cells. Treatment with activating anti-PD-L1 antibodies engaging Fcγ receptors on effector cells leads to the elimination of PD-L1 and p16-positive cells. Our study uncovers a molecular mechanism of p16-dependent regulation of PD-L1 protein stability in senescent cells and reveals the potential of targeting PD-L1 to improve immunosurveillance of senescent cells and ameliorate senescence-associated inflammation.


Asunto(s)
Antígeno B7-H1 , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Estabilidad Proteica , Senescencia Celular/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Animales , Humanos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Vigilancia Inmunológica , Ratones Endogámicos C57BL , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Ratones , Proteolisis , Receptores de IgG/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética
4.
Mamm Genome ; 35(3): 346-361, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39115562

RESUMEN

Pyroptosis has been regarded as caspase-1-mediated monocyte death that induces inflammation, showing a critical and detrimental role in the development of cerebral ischemia-reperfusion injury (IRI). MARCH1 is an E3 ubiquitin ligase that exerts potential anti-inflammatory functions. Therefore, the study probed into the significance of MARCH1 in inflammation and pyroptosis elicited by cerebral IRI. Middle cerebral artery occlusion/reperfusion (MCAO/R)-treated mice and oxygen glucose deprivation/reoxygenation (OGD/R)-treated hippocampal neurons were established to simulate cerebral IRI in vivo and in vitro. MARCH1 and PCSK9 expression was tested in MCAO/R-operated mice, and their interaction was identified by means of the cycloheximide assay and co-immunoprecipitation. The functional roles of MARCH1 and PCSK9 in cerebral IRI were subsequently determined by examining the neurological function, brain tissue changes, neuronal viability, inflammation, and pyroptosis through ectopic expression and knockdown experiments. PCSK9 expression was increased in the brain tissues of MCAO/R mice, while PCSK9 knockdown reduced brain damage and neurological deficits. Additionally, inflammation and pyroptosis were inhibited in OGD/R-exposed hippocampal neurons upon PCSK9 knockdown, accompanied by LDLR upregulation and NLRP3 inflammasome inactivation. Mechanistic experiments revealed that MARCH1 mediated ubiquitination and degradation of PCSK9, lowering PCSK9 protein expression. Furthermore, it was demonstrated that MARCH1 suppressed inflammation and pyroptosis after cerebral IRI by downregulating PCSK9 both in vivo and in vitro. Taken together, the present study demonstrate the protective effect of MARCH1 against cerebral IRI through PCSK9 downregulation, which might contribute to the discovery of new therapies for improving cerebral IRI.


Asunto(s)
Inflamación , Proproteína Convertasa 9 , Piroptosis , Daño por Reperfusión , Ubiquitina-Proteína Ligasas , Animales , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Piroptosis/genética , Proproteína Convertasa 9/genética , Proproteína Convertasa 9/metabolismo , Ratones , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Neuronas/metabolismo , Neuronas/patología , Masculino , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Regulación hacia Abajo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Hipocampo/metabolismo , Hipocampo/patología , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
5.
Nat Commun ; 15(1): 6914, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39134548

RESUMEN

Mitochondrial oxidative phosphorylation (OXPHOS) fuels cellular ATP demands. OXPHOS defects lead to severe human disorders with unexplained tissue specific pathologies. Mitochondrial gene expression is essential for OXPHOS biogenesis since core subunits of the complexes are mitochondrial-encoded. COX14 is required for translation of COX1, the central mitochondrial-encoded subunit of complex IV. Here we describe a COX14 mutant mouse corresponding to a patient with complex IV deficiency. COX14M19I mice display broad tissue-specific pathologies. A hallmark phenotype is severe liver inflammation linked to release of mitochondrial RNA into the cytosol sensed by RIG-1 pathway. We find that mitochondrial RNA release is triggered by increased reactive oxygen species production in the deficiency of complex IV. Additionally, we describe a COA3Y72C mouse, affected in an assembly factor that cooperates with COX14 in early COX1 biogenesis, which displays a similar yet milder inflammatory phenotype. Our study provides insight into a link between defective mitochondrial gene expression and tissue-specific inflammation.


Asunto(s)
Ciclooxigenasa 1 , Complejo IV de Transporte de Electrones , Inflamación , Hígado , Fosforilación Oxidativa , Especies Reactivas de Oxígeno , Animales , Femenino , Humanos , Masculino , Ratones , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/genética , Complejo IV de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/genética , Inflamación/metabolismo , Inflamación/genética , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Proteínas de la Membrana , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Mitocondriales/genética , Mutación , Biosíntesis de Proteínas , Especies Reactivas de Oxígeno/metabolismo , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo
6.
Sci Rep ; 14(1): 19346, 2024 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164324

RESUMEN

Idiopathic orbital inflammation, formerly known as NSOI (nonspecific orbital inflammation), is characterized as a spectrum disorder distinguished by the polymorphic infiltration of lymphoid tissue, presenting a complex and poorly understood etiology. Recent advancements have shed light on the HLF (Human lactoferrin), proposing its critical involvement in the regulation of hematopoiesis and the maintenance of innate mucosal immunity. This revelation has generated significant interest in exploring HLF's utility as a biomarker for NSOI, despite the existing gaps in our understanding of its biosynthetic pathways and operational mechanisms. Intersecting multi-omic datasets-specifically, common differentially expressed genes between GSE58331 and GSE105149 from the Gene Expression Omnibus and immune-related gene compendiums from the ImmPort database-we employed sophisticated analytical methodologies, including Lasso regression and support vector machine-recursive feature elimination, to identify HLF. Gene set enrichment analysis and gene set variation analysis disclosed significant immune pathway enrichment within gene sets linked to HLF. The intricate relationship between HLF expression and immunological processes was further dissected through the utilization of CIBERSORT and ESTIMATE algorithms, which assess characteristics of the immune microenvironment, highlighting a noteworthy association between increased HLF expression and enhanced immune cell infiltration. The expression levels of HLF were corroborated using data from the GSE58331 dataset, reinforcing the validity of our findings. Analysis of 218 HLF-related differentially expressed genes revealed statistically significant discrepancies. Fifteen hub genes were distilled using LASSO and SVM-RFE algorithms. Biological functions connected with HLF, such as leukocyte migration, ossification, and the negative regulation of immune processes, were illuminated. Immune cell analysis depicted a positive correlation between HLF and various cells, including resting mast cells, activated NK cells, plasma cells, and CD8 T cells. Conversely, a negative association was observed with gamma delta T cells, naive B cells, M0 and M1 macrophages, and activated mast cells. Diagnostic assessments of HLF in distinguishing NSOI showed promising accuracy. Our investigation delineates HLF as intricately associated with NSOI, casting light on novel biomarkers for diagnosis and progression monitoring of this perplexing condition.


Asunto(s)
Biología Computacional , Lactoferrina , Aprendizaje Automático , Humanos , Biología Computacional/métodos , Lactoferrina/genética , Lactoferrina/metabolismo , Biomarcadores , Inflamación/genética , Perfilación de la Expresión Génica/métodos , Bases de Datos Genéticas
7.
J Clin Invest ; 134(16)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-39145444

RESUMEN

A disturbed balance between excitation and inhibition (E/I balance) is increasingly recognized as a key driver of neurodegeneration in multiple sclerosis (MS), a chronic inflammatory disease of the central nervous system. To understand how chronic hyperexcitability contributes to neuronal loss in MS, we transcriptionally profiled neurons from mice lacking inhibitory metabotropic glutamate signaling with shifted E/I balance and increased vulnerability to inflammation-induced neurodegeneration. This revealed a prominent induction of the nuclear receptor NR4A2 in neurons. Mechanistically, NR4A2 increased susceptibility to excitotoxicity by stimulating continuous VGF secretion leading to glycolysis-dependent neuronal cell death. Extending these findings to people with MS (pwMS), we observed increased VGF levels in serum and brain biopsies. Notably, neuron-specific deletion of Vgf in a mouse model of MS ameliorated neurodegeneration. These findings underscore the detrimental effect of a persistent metabolic shift driven by excitatory activity as a fundamental mechanism in inflammation-induced neurodegeneration.


Asunto(s)
Glucólisis , Inflamación , Neuronas , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Animales , Ratones , Humanos , Neuronas/metabolismo , Neuronas/patología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/genética , Ratones Noqueados , Transducción de Señal , Masculino , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología
8.
Cell Commun Signal ; 22(1): 399, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143485

RESUMEN

Lipopolysaccharide (LPS)-activated pro-inflammatory responses play a critical role in sepsis, a life-threatening condition. This study investigates the role of origin recognition complex subunit 6 (ORC6) in LPS responses in macrophages and monocytes. Silencing ORC6 using targeted shRNA significantly reduced LPS-induced expression and production of IL-1ß (interleukin-1 beta), TNF-α (tumor necrosis factor alpha), and IL-6 (interleukin-6) in THP-1 human macrophages, peripheral blood mononuclear cells (PBMCs), and bone marrow-derived macrophages (BMDMs). Additionally, ORC6 knockout (KO) via the CRISPR/Cas9 method in THP-1 macrophages inhibited LPS-induced pro-inflammatory responses, while ectopic overexpression of ORC6 enhanced LPS-induced expression and production of pro-inflammatory cytokines. ORC6 is crucial for the activation of the nuclear factor kappa B (NFκB) signaling cascade in macrophages and monocytes. LPS-induced NFκB activation was largely inhibited by ORC6 silencing or KO, but potentiated following ORC6 overexpression. Mechanistically, ORC6 associated with nuclear p65 after LPS stimulation, an interaction necessary for NFκB activation. Overexpression of ORC6 did not recover the reduced pro-inflammatory response to LPS in THP-1 macrophages with silenced p65. Furthermore, the NFκB inhibitor BMS-345,541 nearly eliminated the pro-inflammatory response enhanced by ORC6 overexpression in response to LPS. Further studies revealed that ORC6 depletion inhibited NFκB activation induced by double-stranded RNA (dsRNA) and high mobility group box 1 (HMGB1) in THP-1 macrophages. In vivo experiments demonstrated that macrophage-specific knockdown of ORC6 protected mice from LPS-induced septic shock and inhibited LPS-stimulated production of IL-1ß, TNF-α, and IL-6 in mouse serum. ORC6 silencing also inhibited LPS-induced NFκB activation in ex vivo cultured PBMCs following macrophage-specific knockdown of ORC6. These findings highlight ORC6 as a pivotal mediator in LPS-induced NFκB activation and the pro-inflammatory response in sepsis, suggesting that targeting ORC6 could be a novel therapeutic strategy for managing sepsis and related inflammatory conditions.


Asunto(s)
Inflamación , Lipopolisacáridos , Macrófagos , FN-kappa B , Complejo de Reconocimiento del Origen , Lipopolisacáridos/farmacología , Humanos , Animales , FN-kappa B/metabolismo , Inflamación/metabolismo , Inflamación/genética , Macrófagos/metabolismo , Ratones , Complejo de Reconocimiento del Origen/metabolismo , Complejo de Reconocimiento del Origen/genética , Células THP-1 , Ratones Endogámicos C57BL , Transducción de Señal , Masculino , Monocitos/metabolismo
9.
PLoS One ; 19(8): e0307779, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39150929

RESUMEN

OBJECTIVE: The main pathological change of myocarditis is an inflammatory injury of cardiomyocytes. Long noncoding RNAs (lncRNAs) are closely related to inflammation, and our previous study showed that differential expression of lncRNAs is associated with myocarditis. This study aimed to investigate the impact of lncRNAs on the onset of myocarditis. METHODS: RNA expression was measured by quantitative reverse-transcription polymerase chain reaction (RT-qPCR). Lipopolysaccharide (LPS) was used to induce inflammation in human cardiomyocytes (HCMs). The expression of inflammatory cytokines and myocardial injury markers was detected by enzyme-linked immunosorbent assay (ELISA) and RT-qPCR. Cell viability and apoptosis were measured by the cell counting kit-8 assay and flow cytometry. The binding force between lncRNA NONHSAT122636.2 and microRNA miRNA-2110 was detected using the dual-luciferase assay. RESULTS: NONHSAT122636.2 was dynamically expressed in patients with myocarditis and negatively correlated with inflammation severity. The overexpression of NONHSAT122636.2 improved inflammatory injury in LPS-stimulated HCMs. The study observed that there was a weak binding force between NONHSAT122636.2 and miR-2110. CONCLUSION: NONHSAT122636.2 attenuates myocardial inflammation and apoptosis in myocarditis. Additionally, its expression decreases in the peripheral blood of children suffering from myocarditis and in patients who are diagnosed for the first time showing higher diagnostic sensitivity and specificity. This decrease is negatively correlated with the degree of inflammation. Overall, the study suggests that NONHSAT122636.2 can be exploited as a potential diagnostic biomarker for pediatric myocarditis.


Asunto(s)
Apoptosis , MicroARNs , Miocarditis , Miocitos Cardíacos , ARN Largo no Codificante , Miocarditis/genética , Miocarditis/patología , Miocarditis/metabolismo , ARN Largo no Codificante/genética , Humanos , Apoptosis/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Masculino , Femenino , Lipopolisacáridos/farmacología , Niño , Inflamación/genética , Inflamación/patología , Preescolar , Citocinas/metabolismo , Citocinas/genética
10.
Cell Mol Life Sci ; 81(1): 330, 2024 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-39097839

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a complex syndrome with poorly understood mechanisms driving its early progression (GOLD stages 1-2). Elucidating the genetic factors that influence early-stage COPD, particularly those related to airway inflammation and remodeling, is crucial. This study analyzed lung tissue sequencing data from patients with early-stage COPD (GSE47460) and smoke-exposed mice. We employed Weighted Gene Co-Expression Network Analysis (WGCNA) and machine learning to identify potentially pathogenic genes. Further analyses included single-cell sequencing from both mice and COPD patients to pinpoint gene expression in specific cell types. Cell-cell communication and pseudotemporal analyses were conducted, with findings validated in smoke-exposed mice. Additionally, Mendelian randomization (MR) was used to confirm the association between candidate genes and lung function/COPD. Finally, functional validation was performed in vitro using cell cultures. Machine learning analysis of 30 differentially expressed genes identified 8 key genes, with CLEC5A emerging as a potential pathogenic factor in early-stage COPD. Bioinformatics analyses suggested a role for CLEC5A in macrophage-mediated inflammation during COPD. Two-sample Mendelian randomization linked CLEC5A single nucleotide polymorphisms (SNPs) with Forced Expiratory Volume in One Second (FEV1), FEV1/Forced Vital Capacity (FVC) and early/later on COPD. In vitro, the knockdown of CLEC5A led to a reduction in inflammatory markers within macrophages. Our study identifies CLEC5A as a critical gene in early-stage COPD, contributing to its pathogenesis through pro-inflammatory mechanisms. This discovery offers valuable insights for developing early diagnosis and treatment strategies for COPD and highlights CLEC5A as a promising target for further investigation.


Asunto(s)
Progresión de la Enfermedad , Inflamación , Lectinas Tipo C , Macrófagos , Polimorfismo de Nucleótido Simple , Enfermedad Pulmonar Obstructiva Crónica , Receptores de Superficie Celular , Animales , Humanos , Masculino , Ratones , Inflamación/genética , Inflamación/patología , Inflamación/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Pulmón/patología , Pulmón/metabolismo , Aprendizaje Automático , Macrófagos/metabolismo , Macrófagos/patología , Análisis de la Aleatorización Mendeliana , Ratones Endogámicos C57BL , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
11.
Cell Mol Life Sci ; 81(1): 339, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39120648

RESUMEN

Senataxin is an evolutionarily conserved DNA/RNA helicase, whose dysfunctions are linked to neurodegeneration and cancer. A main activity of this protein is the removal of R-loops, which are nucleic acid structures capable to promote DNA damage and replication stress. Here we found that Senataxin deficiency causes the release of damaged DNA into extranuclear bodies, called micronuclei, triggering the massive recruitment of cGAS, the apical sensor of the innate immunity pathway, and the downstream stimulation of interferon genes. Such cGAS-positive micronuclei are characterized by defective membrane envelope and are particularly abundant in cycling cells lacking Senataxin, but not after exposure to a DNA breaking agent or in absence of the tumor suppressor BRCA1 protein, a partner of Senataxin in R-loop removal. Micronuclei with a discontinuous membrane are normally cleared by autophagy, a process that we show is impaired in Senataxin-deficient cells. The formation of Senataxin-dependent inflamed micronuclei is promoted by the persistence of nuclear R-loops stimulated by the DSIF transcription elongation complex and the engagement of EXO1 nuclease activity on nuclear DNA. Coherently, high levels of EXO1 result in poor prognosis in a subset of tumors lacking Senataxin expression. Hence, R-loop homeostasis impairment, together with autophagy failure and unscheduled EXO1 activity, elicits innate immune response through micronuclei formation in cells lacking Senataxin.


Asunto(s)
Autofagia , Daño del ADN , ADN Helicasas , Inflamación , Enzimas Multifuncionales , Nucleotidiltransferasas , Estructuras R-Loop , ARN Helicasas , Humanos , Autofagia/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/deficiencia , ADN Helicasas/metabolismo , ADN Helicasas/genética , ADN Helicasas/deficiencia , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/deficiencia , Exodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/genética , Inmunidad Innata , Inflamación/patología , Inflamación/metabolismo , Inflamación/genética , Enzimas Multifuncionales/metabolismo , Enzimas Multifuncionales/genética , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/genética , Fosfoproteínas , ARN Helicasas/metabolismo , ARN Helicasas/genética
12.
Arch Biochem Biophys ; 759: 110112, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39111613

RESUMEN

Inflammation is the body's response to injuries, which depends on numerous regulatory factors. Among them, miRNAs have gained much attention for their role in regulating inflammatory gene expression at multiple levels. In particular, miR-21 is up-regulated during the inflammatory response and reported to be involved in the resolution of inflammation by down-regulating pro-inflammatory mediators, including MyD88. Herein, we evaluated the regulatory effects of miR-21 on the TLR-4/MyD88 pathway in an in vitro model of 6-mer HA oligosaccharides-induced inflammation in human chondrocytes. The exposition of chondrocytes to 6-mer HA induced the activation of the TLR4/MyD88 pathway, which culminates in NF-kB activation. Changes in miR-21, TLR-4, MyD88, NLRP3 inflammasome, IL-29, Caspase1, MMP-9, iNOS, and COX-2 mRNA expression of 6-mer HA-stimulated chondrocytes were examined by qRT-PCR. Protein amounts of TLR-4, MyD88, NLRP3 inflammasome, p-ERK1/2, p-AKT, IL-29, caspase1, MMP-9, p-NK-kB p65 subunit, and IKB-a have been evaluated by ELISA kits. NO and PGE2 levels have been assayed by colorimetric and ELISA kits, respectively. HA oligosaccharides induced a significant increase in the expression of the above parameters, including NF-kB activity. The use of a miR-21 mimic attenuated MyD88 expression levels and the downstream effectors. On the contrary, treatment with a miR-21 inhibitor induced opposite effects. Interestingly, the use of a MyD88 siRNA confirmed MyD88 as the target of miR-21 action. Our results suggest that miR-21 expression could increase in an attempt to reduce the inflammatory response, targeting MyD88.


Asunto(s)
Condrocitos , Ácido Hialurónico , Inflamación , MicroARNs , Factor 88 de Diferenciación Mieloide , Oligosacáridos , Humanos , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética , MicroARNs/genética , MicroARNs/metabolismo , Condrocitos/metabolismo , Condrocitos/efectos de los fármacos , Ácido Hialurónico/farmacología , Ácido Hialurónico/metabolismo , Inflamación/metabolismo , Inflamación/genética , Oligosacáridos/farmacología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Transducción de Señal/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , FN-kappa B/metabolismo , Células Cultivadas
13.
Gene ; 929: 148812, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39116959

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and represents the main cause of liver cirrhosis and hepatocellular carcinoma. Cav3.2 is a T-type calcium channel that is widely present in tissues throughout the body and plays a vital role in energy and metabolic balance. However, the effects of Cav3.2 on the NFALD remain unclear. Here, we investigated the role of Cav3.2 channel in the development and progression of NAFLD. After 16 weeks on a high-fat diets (HFD), Cav3.2 knockout (Cav3.2 KO) improved hepatic steatosis, liver injury and metabolic syndrome in an NAFLD mouse model. We provided evidence that Cav3.2 KO inhibited HFD-induced hepatic oxidative stress, inflammation and hepatocyte apoptosis. In addition, Cav3.2 KO also attenuated hepatic lipid accumulation, oxidative stress, inflammation and hepatocyte apoptosis in palmitic acid/oleic acid (PAOA)-treated primary hepatocytes. These results suggest that therapeutic approaches targeting Cav3.2 provide effective approaches for treating NAFLD.


Asunto(s)
Apoptosis , Canales de Calcio Tipo T , Dieta Alta en Grasa , Hepatocitos , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , Estrés Oxidativo , Animales , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Canales de Calcio Tipo T/genética , Canales de Calcio Tipo T/metabolismo , Ratones , Dieta Alta en Grasa/efectos adversos , Hepatocitos/metabolismo , Hepatocitos/patología , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Hígado/metabolismo , Hígado/patología , Inflamación/genética , Inflamación/metabolismo
14.
Proc Natl Acad Sci U S A ; 121(34): e2319724121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39141348

RESUMEN

Skeletal muscle atrophy is a morbidity and mortality risk factor that happens with disuse, chronic disease, and aging. The tissue remodeling that happens during recovery from atrophy or injury involves changes in different cell types such as muscle fibers, and satellite and immune cells. Here, we show that the previously uncharacterized gene and protein Zfp697 is a damage-induced regulator of muscle remodeling. Zfp697/ZNF697 expression is transiently elevated during recovery from muscle atrophy or injury in mice and humans. Sustained Zfp697 expression in mouse muscle leads to a gene expression signature of chemokine secretion, immune cell recruitment, and extracellular matrix remodeling. Notably, although Zfp697 is expressed in several cell types in skeletal muscle, myofiber-specific Zfp697 genetic ablation in mice is sufficient to hinder the inflammatory and regenerative response to muscle injury, compromising functional recovery. We show that Zfp697 is an essential mediator of the interferon gamma response in muscle cells and that it functions primarily as an RNA-interacting protein, with a very high number of miRNA targets. This work identifies Zfp697 as an integrator of cell-cell communication necessary for tissue remodeling and regeneration.


Asunto(s)
Músculo Esquelético , Proteínas de Unión al ARN , Animales , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Ratones Noqueados , Atrofia Muscular/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , MicroARNs/genética , MicroARNs/metabolismo , Ratones Endogámicos C57BL , Interferón gamma/metabolismo
15.
Mol Biol Rep ; 51(1): 909, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39145884

RESUMEN

BACKGROUND: Inflammatory cytokines such as Interleukin 1ß(IL1ß), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS: High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS: Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.


Asunto(s)
Diferenciación Celular , Redes Reguladoras de Genes , Osteogénesis , ARN Endógeno Competitivo , ARN Largo no Codificante , Factor de Necrosis Tumoral alfa , Animales , Humanos , Ratones , Apoptosis/genética , Diferenciación Celular/genética , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/genética , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Osteogénesis/genética , ARN Endógeno Competitivo/genética , ARN Endógeno Competitivo/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Funct Integr Genomics ; 24(5): 140, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39160285

RESUMEN

Acute pancreatitis (AP) is an inflammatory disease of the pancreas and the main cause of hospital admissions for gastrointestinal diseases. Here, the work studied the circular RNA DTNB/microRNA-485-5p/MCL1 axis in AP and hoped to unravel the related mechanism. Caerulein exposure replicated an AP model in AR42J cells, and caerulein-mediated expression of circDTNB, miR-485-5p, and MCL1 was recorded. After exposure, cells were intervened with transfection plasmids and tested for LDH release, apoptosis, and inflammation. To determine the interwork of circDTNB, miR-485-5p, and MCL1, prediction results and verification experiments were conducted. Caerulein exposure reduced circDTNB and MCL1, while elevated miR-485-5p levels in AR42J cells. Upregulating circDTNB protected AR42J cells from caerulein-induced LDH cytotoxicity, apoptosis, and inflammation, but circDTNB upregulation-induced protections could be muffled by inhibiting MCL1. On the contrary, downregulating circDTNB further damaged AR42J cells under caerulein exposure, however, this phenomenon could be partially rescued after silencing miR-485-5p. miR-485-5p was mechanistically verified to be a target of circDTNB to mediate MCL1. Overall, the circDTNB/miR-485-5p/MCL1 axis protects inflammatory response and apoptosis in caerulein-exposed AR42J cells, promisingly identifying circDTNB as a novel molecule for AP treatment.


Asunto(s)
Apoptosis , Ceruletida , Inflamación , MicroARNs , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , ARN Circular , MicroARNs/genética , MicroARNs/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Animales , Ratas , Inflamación/genética , Pancreatitis/genética , Pancreatitis/metabolismo , Pancreatitis/inducido químicamente , Pancreatitis/patología , Línea Celular
17.
Int J Mol Sci ; 25(15)2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39125844

RESUMEN

TNF-α-induced protein 3 (TNFAIP3), commonly referred to as A20, is an integral part of the ubiquitin-editing complex that significantly influences immune regulation, apoptosis, and the initiation of diverse immune responses. The A20 protein is characterized by an N-terminal ovarian tumor (OTU) domain and a series of seven zinc finger (ZNF) domains. Mutations in the TNFAIP3 gene are implicated in various immune-related diseases, such as Behçet's disease, polyarticular juvenile idiopathic arthritis, autoimmune thyroiditis, autoimmune hepatitis, and rheumatoid arthritis. These mutations can lead to a spectrum of symptoms, including, but not limited to, recurrent fever, ulcers, rashes, musculoskeletal and gastrointestinal dysfunctions, cardiovascular issues, and respiratory infections. The majority of these mutations are either nonsense (STOP codon) or frameshift mutations, which are typically associated with immune dysfunctions. Nonetheless, missense mutations have also been identified as contributors to these conditions. These genetic alterations may interfere with several biological pathways, notably abnormal NF-κB signaling and dysregulated ubiquitination. Currently, there is no definitive treatment for A20 haploinsufficiency; however, therapeutic strategies can alleviate the symptoms in patients. This review delves into the mutations reported in the TNFAIP3 gene, the clinical progression in affected individuals, potential disease mechanisms, and a brief overview of the available pharmacological interventions for A20 haploinsufficiency. Mandatory genetic testing of the TNFAIP3 gene should be performed in patients diagnosed with autoinflammatory disorders to better understand the genetic underpinnings and guide treatment decisions.


Asunto(s)
Haploinsuficiencia , Mutación , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Humanos , Haploinsuficiencia/genética , Inflamación/genética , Predisposición Genética a la Enfermedad , Animales
18.
Cell Death Dis ; 15(8): 574, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117638

RESUMEN

Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNß, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNß-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNß, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.


Asunto(s)
Citocinas , Péptidos y Proteínas de Señalización Intracelular , Necroptosis , Proteínas Serina-Treonina Quinasas , Tristetraprolina , Tristetraprolina/metabolismo , Tristetraprolina/genética , Animales , Citocinas/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Macrófagos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/genética , Células RAW 264.7 , Regulación hacia Arriba/efectos de los fármacos , Interferón beta/metabolismo , Ratones Endogámicos C57BL , Regulación de la Expresión Génica
19.
Clin Exp Hypertens ; 46(1): 2390419, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-39133866

RESUMEN

BACKGROUND: Complex interconnections are evident among gut microbiota, circulating metabolites, inflammatory cytokines, and the pathogenesis of abdominal aortic aneurysms (AAA), with the causal dynamics yet to be comprehensively elucidated. The primary objective of this study was to elucidate the potential causal relationships involving gut microbiota-mediated plasma metabolites, inflammatory cytokines, and AAA. METHODS: We utilized data from genome-wide association studies predominantly comprising individuals of European ancestry, encompassing four major gut microbiota signatures, 233 plasma metabolite signatures (N = 136,016), 91 inflammatory cytokine signatures (N = 14,824), and AAA signatures (N = 1,458,875). Mendelian randomization (MR), employed in a two-sample format, was utilized as a tool to investigate the potential causal pathways from gut microbiota to the development of AAA. Additionally, a two-step MR approach was employed to dissect the impact of plasma metabolites and inflammatory cytokines on the relationship between gut microbiota and AAA and to ascertain the mediated fractions. RESULTS: Our findings indicate that five phylum or family-identical bacteria, 175 plasma metabolites, and seven inflammatory factors are causally associated with AAA. Among them, five bacterial species from the same phylum or family, identified from different GWAS data, were strongly associated with AAA. Of these, two exhibited negative causality and three exhibited positive causality. We found that the phylum Firmicutes and the families Oscillospiraceae might reduce the risk of AAA, whereas the families Prevotellaceae, Sutterellaceae, and Aminobacteriaceae might increase the risk of AAA. Further screening indicated that phylum Firmicutes id.1672 (GCST90017114) may confer a protective effect against AAA by reducing triglyceride levels in medium/small high-density lipoprotein (HDL). CONCLUSION: MR analysis has delineated a causal pathway from gut microbiota, through plasma circulating metabolites and inflammatory cytokines, to the pathogenesis of AAA. The role of intestinal flora and certain biomarkers may provide a reference for the diagnosis of AAA, and contribute to the prevention, diagnosis, and treatment of AAA disease.


Asunto(s)
Aneurisma de la Aorta Abdominal , Citocinas , Microbioma Gastrointestinal , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Aneurisma de la Aorta Abdominal/microbiología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/genética , Humanos , Microbioma Gastrointestinal/fisiología , Microbioma Gastrointestinal/genética , Citocinas/sangre , Masculino , Femenino , Inflamación/sangre , Inflamación/genética
20.
Brain Behav ; 14(8): e3615, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39135276

RESUMEN

BACKGROUND: Temporal lobe epilepsy (TLE), a prevalent neurological disorder, is associated with hippocampal oxidative stress and inflammation. A recent study reveals that the long noncoding RNA ILF3 divergent transcript (ILF3-AS1) level is elevated in the hippocampus of TLE patients; however, the functional roles of ILF3-AS1 in TLE and underlying mechanisms deserve further investigation. Hence, this study aimed to elucidate whether ILF3-AS1 is involved in the pathogenesis of TLE by regulating oxidative stress and inflammation and to explore its underlying mechanism in vitro. METHODS: Human hippocampal neurons were subjected to a magnesium-free (Mg2+-free) solution to establish an in vitro model of TLE. The potential binding sites between ILF3-AS1 and miRNA were predicted by TargetScan/Starbase and confirmed by dual luciferase reporter assay. Cell viability and damage were assessed by cell counting kit-8 and lactate dehydrogenase assay kits, respectively. Levels of reactive oxygen species, malondialdehyde, and superoxide dismutase were determined by commercial kits. Levels of Interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-alpha were quantified by enzyme-linked immunosorbent assay. The expressions of gene and protein were determined by quantitative real-time polymerase chain reaction and Western blot analysis. RESULTS: In Mg2+-free-treated hippocampal neurons, both ILF3-AS1 and HMGB1 were highly up-regulated, whereas miR-504-3p was down-regulated. ILF3-AS1 knockdown ameliorated Mg2+-free-induced cellular damage, oxidative stress, and inflammatory response. Bioinformatics analysis revealed that miR-504-3p was a target of ILF3-AS1 and was negatively regulated by ILF3-AS1. MiR-504-3p inhibitor blocked the protection of ILF3-AS1 knockdown against Mg2+-free-induced neuronal injury. Further analysis presented that ILF3-AS1 regulated HMGB1 expression by sponging miR-504-3p. Moreover, HMGB1 overexpression reversed the protective functions of ILF3-AS1 knockdown. CONCLUSION: Our findings indicate that ILF3-AS1 contributes to Mg2+-free-induced hippocampal neuron injuries, oxidative stress, and inflammation by targeting the miR-504-3p/HMGB1 axis. These results provide a novel mechanistic understanding of ILF3-AS1 in TLE and suggest potential therapeutic targets for the treatment of epilepsy.


Asunto(s)
Epilepsia del Lóbulo Temporal , Proteína HMGB1 , Hipocampo , Inflamación , MicroARNs , Estrés Oxidativo , ARN Largo no Codificante , Estrés Oxidativo/fisiología , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Inflamación/genética , Neuronas/metabolismo , Proteínas del Factor Nuclear 90/metabolismo , Proteínas del Factor Nuclear 90/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA