Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Viruses ; 15(3)2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36992368

RESUMEN

The importance of genomic surveillance on emerging diseases continues to be highlighted with the ongoing SARS-CoV-2 pandemic. Here, we present an analysis of a new bat-borne mumps virus (MuV) in a captive colony of lesser dawn bats (Eonycteris spelaea). This report describes an investigation of MuV-specific data originally collected as part of a longitudinal virome study of apparently healthy, captive lesser dawn bats in Southeast Asia (BioProject ID PRJNA561193) which was the first report of a MuV-like virus, named dawn bat paramyxovirus (DbPV), in bats outside of Africa. More in-depth analysis of these original RNA sequences in the current report reveals that the new DbPV genome shares only 86% amino acid identity with the RNA-dependent RNA polymerase of its closest relative, the African bat-borne mumps virus (AbMuV). While there is no obvious immediate cause for concern, it is important to continue investigating and monitoring bat-borne MuVs to determine the risk of human infection.


Asunto(s)
COVID-19 , Quirópteros , Animales , Humanos , Virus de la Parotiditis/genética , Filogenia , SARS-CoV-2 , Genómica , Asia Sudoriental/epidemiología , Paramyxoviridae/genética
4.
Sci Rep ; 12(1): 3463, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35236896

RESUMEN

Early detection of diseases such as COVID-19 could be a critical tool in reducing disease transmission by helping individuals recognize when they should self-isolate, seek testing, and obtain early medical intervention. Consumer wearable devices that continuously measure physiological metrics hold promise as tools for early illness detection. We gathered daily questionnaire data and physiological data using a consumer wearable (Oura Ring) from 63,153 participants, of whom 704 self-reported possible COVID-19 disease. We selected 73 of these 704 participants with reliable confirmation of COVID-19 by PCR testing and high-quality physiological data for algorithm training to identify onset of COVID-19 using machine learning classification. The algorithm identified COVID-19 an average of 2.75 days before participants sought diagnostic testing with a sensitivity of 82% and specificity of 63%. The receiving operating characteristic (ROC) area under the curve (AUC) was 0.819 (95% CI [0.809, 0.830]). Including continuous temperature yielded an AUC 4.9% higher than without this feature. For further validation, we obtained SARS CoV-2 antibody in a subset of participants and identified 10 additional participants who self-reported COVID-19 disease with antibody confirmation. The algorithm had an overall ROC AUC of 0.819 (95% CI [0.809, 0.830]), with a sensitivity of 90% and specificity of 80% in these additional participants. Finally, we observed substantial variation in accuracy based on age and biological sex. Findings highlight the importance of including temperature assessment, using continuous physiological features for alignment, and including diverse populations in algorithm development to optimize accuracy in COVID-19 detection from wearables.


Asunto(s)
Temperatura Corporal , COVID-19/diagnóstico , Dispositivos Electrónicos Vestibles , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , COVID-19/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , SARS-CoV-2/aislamiento & purificación , Adulto Joven
5.
Sci Rep ; 10(1): 11506, 2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32661418

RESUMEN

Helicobacter pylori is a gram-negative bacterium that persistently colonizes the human stomach by inducing immunoregulatory responses. We have used a novel platform that integrates a bone marrow-derived macrophage and live H. pylori co-culture with global time-course transcriptomics analysis to identify new regulatory genes based on expression patterns resembling those of genes with known regulatory function. We have used filtering criteria based on cellular location and novelty parameters to select 5 top lead candidate targets. Of these, Plexin domain containing 2 (Plxdc2) was selected as the top lead immunoregulatory target. Loss of function studies with in vivo models of H. pylori infection as well as a chemically-induced model of colitis, confirmed its predicted regulatory function and significant impact on modulation of the host immune response. Our integrated bioinformatics analyses and experimental validation platform has enabled the discovery of new immunoregulatory genes. This pipeline can be used for the identification of genes with therapeutic applications for treating infectious, inflammatory, and autoimmune diseases.


Asunto(s)
Genes Reguladores/genética , Infecciones por Helicobacter/genética , Helicobacter pylori/genética , Macrófagos/metabolismo , Animales , Técnicas de Cocultivo , Simulación por Computador , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/patogenicidad , Humanos , Macrófagos/microbiología , Ratones , RNA-Seq , Receptores de Superficie Celular/genética
6.
Viruses ; 12(5)2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32422932

RESUMEN

Rousettus bat coronavirus GCCDC1 (RoBat-CoV GCCDC1) is a cross-family recombinant coronavirus that has previously only been reported in wild-caught bats in Yúnnan, China. We report the persistence of a related strain in a captive colony of lesser dawn bats captured in Singapore. Genomic evidence of the virus was detected using targeted enrichment sequencing, and further investigated using deeper, unbiased high throughput sequencing. RoBat-CoV GCCDC1 Singapore shared 96.52% similarity with RoBat-CoV GCCDC1 356 (NC_030886) at the nucleotide level, and had a high prevalence in the captive bat colony. It was detected at five out of six sampling time points across the course of 18 months. A partial segment 1 from an ancestral Pteropine orthoreovirus, p10, makes up the recombinant portion of the virus, which shares high similarity with previously reported RoBat-CoV GCCDC1 strains that were detected in Yúnnan, China. RoBat-CoV GCCDC1 is an intriguing, cross-family recombinant virus, with a geographical range that expands farther than was previously known. The discovery of RoBat-CoV GCCDC1 in Singapore indicates that this recombinant coronavirus exists in a broad geographical range, and can persist in bat colonies long-term.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Quirópteros/virología , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/veterinaria , Animales , Betacoronavirus/genética , Reservorios de Enfermedades/virología , Genoma Viral/genética , Geografía , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia , Recombinación Genética/genética , Singapur/epidemiología
7.
Virus Evol ; 6(1): veaa017, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33747541

RESUMEN

The virosphere is largely unexplored and the majority of viruses are yet to be represented in public sequence databases. Bats are rich reservoirs of viruses, including several zoonoses. In this study, high throughput sequencing (HTS) of viral RNA extracted from swabs of four body sites per bat per timepoint is used to characterize the virome through a longitudinal study of a captive colony of fruit nectar bats, species Eonycteris spelaea in Singapore. Through unbiased shotgun and target enrichment sequencing, we identify both known and previously unknown viruses of zoonotic relevance and define the population persistence and temporal patterns of viruses from families that have the capacity to jump the species barrier. To our knowledge, this is the first study that combines probe-based viral enrichment with HTS to create a viral profile from multiple swab sites on individual bats and their cohort. This work demonstrates temporal patterns of the lesser dawn bat virome, including several novel viruses. Given the known risk for bat-human zoonoses, a more complete understanding of the viral dynamics in South-eastern Asian bats has significant implications for disease prevention and control. The findings of this study will be of interest to U.S. Department of Defense personnel stationed in the Asia-Pacific region and regional public health laboratories engaged in emerging infectious disease surveillance efforts.

8.
BMC Infect Dis ; 19(1): 905, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31660864

RESUMEN

BACKGROUND: Antibiotic resistance is rising at disturbing rates and contributes to the deaths of millions of people yearly. Antibiotic resistant infections disproportionately affect those with immunocompromising conditions, chronic colonization, and frequent antibiotic use such as transplant patients or those with cystic fibrosis. However, clinicians lack the diagnostic tools to confidently diagnose and treat infections, leading to widespread use of empiric broad spectrum antimicrobials, often for prolonged duration. CASE PRESENTATION: A 22 year-old Caucasian female with cystic fibrosis received a bilateral orthotopic lung transplantation 5 months prior to the index hospitalization. She underwent routine surveillance bronchoscopy and was admitted for post-procedure fever. A clear cause of infection was not identified by routine methods. Imaging and bronchoscopic lung biopsy did not identify an infectious agent or rejection. She was treated with a prolonged course of antimicrobials targeting known colonizing organisms from prior bronchoalveolar lavage cultures (Pseudomonas, Staphylococcus aureus, and Aspergillus). However, we identified Stenotrophomonas maltophilia in two independent whole blood samples using direct-pathogen sequencing, which was not identified by other methods. CONCLUSIONS: This case represents a common clinical conundrum: identification of infection in a high-risk, complex patient. Here, direct-pathogen sequencing identified a pathogen that would not otherwise have been identified by common techniques. Had results been clinically available, treatment could have been customized, avoiding a prolonged course of broad spectrum antimicrobials that would only exacerbate resistance. Direct-pathogen sequencing is poised to fill a diagnostic gap for pathogen identification, allowing early identification and customization of treatment in a culture-independent, pathogen-agnostic manner.


Asunto(s)
Broncoscopía/efectos adversos , Fiebre/etiología , Infecciones por Bacterias Gramnegativas/diagnóstico , Infecciones por Bacterias Gramnegativas/etiología , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis de Secuencia de ARN , Stenotrophomonas maltophilia/genética , Antibacterianos/uso terapéutico , Lavado Broncoalveolar , Toma de Decisiones Clínicas , Fibrosis Quística/cirugía , Farmacorresistencia Bacteriana , Femenino , Fiebre/tratamiento farmacológico , Humanos , Trasplante de Pulmón , Pseudomonas/aislamiento & purificación , Staphylococcus aureus/aislamiento & purificación , Resultado del Tratamiento , Adulto Joven
9.
Viruses ; 10(4)2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29642590

RESUMEN

Multi-drug resistance is increasing at alarming rates. The efficacy of phage therapy, treating bacterial infections with bacteriophages alone or in combination with traditional antibiotics, has been demonstrated in emergency cases in the United States and in other countries, however remains to be approved for wide-spread use in the US. One limiting factor is a lack of guidelines for assessing the genomic safety of phage candidates. We present the phage characterization workflow used by our team to generate data for submitting phages to the Federal Drug Administration (FDA) for authorized use. Essential analysis checkpoints and warnings are detailed for obtaining high-quality genomes, excluding undesirable candidates, rigorously assessing a phage genome for safety and evaluating sequencing contamination. This workflow has been developed in accordance with community standards for high-throughput sequencing of viral genomes as well as principles for ideal phages used for therapy. The feasibility and utility of the pipeline is demonstrated on two new phage genomes that meet all safety criteria. We propose these guidelines as a minimum standard for phages being submitted to the FDA for review as investigational new drug candidates.


Asunto(s)
Bacteriófagos/genética , Genoma Viral/genética , Terapia de Fagos/normas , Bacteriófagos/clasificación , Bacteriófagos/aislamiento & purificación , Bacteriófagos/fisiología , Genómica , Guías como Asunto , Humanos , Filogenia , Reproducibilidad de los Resultados , Flujo de Trabajo
10.
J Immunol ; 198(8): 3195-3204, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28264969

RESUMEN

Helicobacter pylori, the dominant member of the human gastric microbiota, elicits immunoregulatory responses implicated in protective versus pathological outcomes. To evaluate the role of macrophages during infection, we employed a system with a shifted proinflammatory macrophage phenotype by deleting PPARγ in myeloid cells and found a 5- to 10-fold decrease in gastric bacterial loads. Higher levels of colonization in wild-type mice were associated with increased presence of mononuclear phagocytes and in particular with the accumulation of CD11b+F4/80hiCD64+CX3CR1+ macrophages in the gastric lamina propria. Depletion of phagocytic cells by clodronate liposomes in wild-type mice resulted in a reduction of gastric H. pylori colonization compared with nontreated mice. PPARγ-deficient and macrophage-depleted mice presented decreased IL-10-mediated myeloid and T cell regulatory responses soon after infection. IL-10 neutralization during H. pylori infection led to increased IL-17-mediated responses and increased neutrophil accumulation at the gastric mucosa. In conclusion, we report the induction of IL-10-driven regulatory responses mediated by CD11b+F4/80hiCD64+CX3CR1+ mononuclear phagocytes that contribute to maintaining high levels of H. pylori loads in the stomach by modulating effector T cell responses at the gastric mucosa.


Asunto(s)
Mucosa Gástrica/inmunología , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/inmunología , Macrófagos/inmunología , Animales , Modelos Animales de Enfermedad , Citometría de Flujo , Helicobacter pylori , Ratones , Ratones Endogámicos C57BL
11.
Oncotarget ; 7(22): 33096-110, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27105514

RESUMEN

Histiocytic sarcoma is an uncommon malignancy in both humans and veterinary species. Research exploring the pathogenesis of this disease is scarce; thus, diagnostic and therapeutic options for patients are limited. Recent publications have suggested a role for the NLR, NLRX1, in acting as a tumor suppressor. Based on these prior findings, we hypothesized that NLRX1 would function to inhibit tumorigenesis and thus the development of histiocytic sarcoma. To test this, we utilized Nlrx1-/- mice and a model of urethane-induced tumorigenesis. Nlrx1-/- mice exposed to urethane developed splenic histiocytic sarcoma that was associated with significant up-regulation of the NF-κB signaling pathway. Additionally, development of these tumors was also significantly associated with the increased regulation of genes associated with AKT signaling, cell death and autophagy. Together, these data show that NLRX1 suppresses tumorigenesis and reveals new genetic pathways involved in the pathobiology of histiocytic sarcoma.


Asunto(s)
Sarcoma Histiocítico/metabolismo , Proteínas Mitocondriales/metabolismo , FN-kappa B/metabolismo , Animales , Carcinogénesis , Modelos Animales de Enfermedad , Femenino , Sarcoma Histiocítico/genética , Sarcoma Histiocítico/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/genética , FN-kappa B/genética , Transducción de Señal
12.
PLoS One ; 10(9): e0137839, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26367386

RESUMEN

Helicobacter pylori colonizes half of the world's population as the dominant member of the gastric microbiota resulting in a lifelong chronic infection. Host responses toward the bacterium can result in asymptomatic, pathogenic or even favorable health outcomes; however, mechanisms underlying the dual role of H. pylori as a commensal versus pathogenic organism are not well characterized. Recent evidence suggests mononuclear phagocytes are largely involved in shaping dominant immunity during infection mediating the balance between host tolerance and succumbing to overt disease. We combined computational modeling, bioinformatics and experimental validation in order to investigate interactions between macrophages and intracellular H. pylori. Global transcriptomic analysis on bone marrow-derived macrophages (BMDM) in a gentamycin protection assay at six time points unveiled the presence of three sequential host response waves: an early transient regulatory gene module followed by sustained and late effector responses. Kinetic behaviors of pattern recognition receptors (PRRs) are linked to differential expression of spatiotemporal response waves and function to induce effector immunity through extracellular and intracellular detection of H. pylori. We report that bacterial interaction with the host intracellular environment caused significant suppression of regulatory NLRC3 and NLRX1 in a pattern inverse to early regulatory responses. To further delineate complex immune responses and pathway crosstalk between effector and regulatory PRRs, we built a computational model calibrated using time-series RNAseq data. Our validated computational hypotheses are that: 1) NLRX1 expression regulates bacterial burden in macrophages; and 2) early host response cytokines down-regulate NLRX1 expression through a negative feedback circuit. This paper applies modeling approaches to characterize the regulatory role of NLRX1 in mechanisms of host tolerance employed by macrophages to respond to and/or to co-exist with intracellular H. pylori.


Asunto(s)
Infecciones por Helicobacter/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/fisiología , Proteínas Mitocondriales/metabolismo , Animales , Células Cultivadas , Simulación por Computador , Femenino , Regulación de la Expresión Génica , Infecciones por Helicobacter/microbiología , Helicobacter pylori/patogenicidad , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Proteínas Mitocondriales/genética , Modelos Biológicos
13.
Curr Drug Targets ; 15(6): 565-72, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24628287

RESUMEN

Lanthionine synthetase component C-like protein 2 (LANCL2) is a member of the LANCL protein family, which is broadly expressed throughout the body. LANCL2 is the molecular target of abscisic acid (ABA), a compound with insulin-sensitizing and immune modulatory actions. LANCL2 is required for membrane binding and signaling of ABA in immune cells. Direct binding of ABA to LANCL2 was predicted in silico using molecular modeling approaches and validated experimentally using ligand-binding assays and kinetic surface plasmon resonance studies. The therapeutic potential of the LANCL2 pathway ranges from increasing cellular sensitivity to anticancer drugs, insulin-sensitizing effects and modulating immune and inflammatory responses in the context of immune-mediated and infectious diseases. A case for LANCL2-based drug discovery and development is also illustrated by the anti-inflammatory activity of novel LANCL2 ligands such as NSC61610 against inflammatory bowel disease and influenza-driven inflammation in mice. This review discusses the value of LANCL2 as a novel therapeutic target for the discovery and development of new classes of orally active drugs against chronic metabolic, immune-mediated and infectious diseases.


Asunto(s)
Antiinflamatorios/farmacología , Inflamación/tratamiento farmacológico , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Ácido Abscísico/metabolismo , Animales , Humanos , Hipoglucemiantes/farmacología , Inflamación/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Modelos Moleculares , Proteínas de Unión a Fosfato , Conformación Proteica
14.
Gut Microbes ; 4(4): 281-91, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23680797

RESUMEN

Enteroaggregative Escherichia coli (EAEC) has been acknowledged as an emerging cause of gastroenteritis worldwide for over two decades. Epidemiologists are revealing the role of EAEC in diarrheal outbreaks as a more common occurrence than ever suggested before. EAEC induced diarrhea is most commonly associated with travelers, children and immunocompromised individuals however its afflictions are not limited to any particular demographic. Many attributes have been discovered and characterized surrounding the capability of EAEC to provoke a potent pro-inflammatory immune response, however cellular and molecular mechanisms underlying initiation, progression and outcomes are largely unknown. This limited understanding can be attributed to heterogeneity in strains and the lack of adequate animal models. This review aims to summarize current knowledge about EAEC etiology, pathogenesis and clinical manifestation. Additionally, current animal models and their limitations will be discussed along with the value of applying systems-wide approaches such as computational modeling to study host-EAEC interactions.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Escherichia coli/patogenicidad , Gastroenteritis/microbiología , Gastroenteritis/patología , Animales , Diarrea/microbiología , Diarrea/patología , Humanos
15.
PLoS Comput Biol ; 9(4): e1003027, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23592971

RESUMEN

Differentiation of CD4+ T cells into effector or regulatory phenotypes is tightly controlled by the cytokine milieu, complex intracellular signaling networks and numerous transcriptional regulators. We combined experimental approaches and computational modeling to investigate the mechanisms controlling differentiation and plasticity of CD4+ T cells in the gut of mice. Our computational model encompasses the major intracellular pathways involved in CD4+ T cell differentiation into T helper 1 (Th1), Th2, Th17 and induced regulatory T cells (iTreg). Our modeling efforts predicted a critical role for peroxisome proliferator-activated receptor gamma (PPARγ) in modulating plasticity between Th17 and iTreg cells. PPARγ regulates differentiation, activation and cytokine production, thereby controlling the induction of effector and regulatory responses, and is a promising therapeutic target for dysregulated immune responses and inflammation. Our modeling efforts predict that following PPARγ activation, Th17 cells undergo phenotype switch and become iTreg cells. This prediction was validated by results of adoptive transfer studies showing an increase of colonic iTreg and a decrease of Th17 cells in the gut mucosa of mice with colitis following pharmacological activation of PPARγ. Deletion of PPARγ in CD4+ T cells impaired mucosal iTreg and enhanced colitogenic Th17 responses in mice with CD4+ T cell-induced colitis. Thus, for the first time we provide novel molecular evidence in vivo demonstrating that PPARγ in addition to regulating CD4+ T cell differentiation also plays a major role controlling Th17 and iTreg plasticity in the gut mucosa.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Biología Computacional/métodos , Citocinas/metabolismo , Animales , Diferenciación Celular , Simulación por Computador , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Modelos Moleculares , Modelos Teóricos , PPAR gamma/metabolismo , Fenotipo , Transducción de Señal , Células Th17/metabolismo
16.
PLoS One ; 8(2): e57812, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469071

RESUMEN

BACKGROUND: Enteroaggregative Escherichia coli (EAEC) is recognized as an emerging cause of persistent diarrhea and enteric disease worldwide. Mucosal immunity towards EAEC infections is incompletely understood due in part to the lack of appropriate animal models. This study presents a new mouse model and investigates the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the modulation of host responses to EAEC in nourished and malnourished mice. METHODS/PRINCIPAL FINDINGS: Wild-type and T cell-specific PPARγ null C57BL/6 mice were fed protein-deficient diets at weaning and challenged with 5×10(9)cfu EAEC strain JM221 to measure colonic gene expression and immune responses to EAEC. Antigen-specific responses to E. coli antigens were measured in nourished and malnourished mice following infection and demonstrated the immunosuppressive effects of malnutrition at the cellular level. At the molecular level, both pharmacological blockade and deletion of PPARγ in T cells resulted in upregulation of TGF-ß, IL-6, IL-17 and anti-microbial peptides, enhanced Th17 responses, fewer colonic lesions, faster clearance of EAEC, and improved recovery. The beneficial effects of PPARγ blockade on weight loss and EAEC clearance were abrogated by neutralizing IL-17 in vivo. CONCLUSIONS: Our studies provide in vivo evidence supporting the beneficial role of mucosal innate and effector T cell responses on EAEC burden and suggest pharmacological blockade of PPARγ as a novel therapeutic intervention for EAEC infection.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Escherichia coli/fisiología , PPAR gamma/metabolismo , Anilidas/farmacología , Animales , Antígenos Bacterianos/inmunología , Carga Bacteriana/genética , Carga Bacteriana/inmunología , Peso Corporal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Escherichia coli/inmunología , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Técnicas de Inactivación de Genes , Interleucina-17/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Desnutrición/complicaciones , Ratones , Ratones Endogámicos C57BL , PPAR gamma/antagonistas & inhibidores , PPAR gamma/deficiencia , PPAR gamma/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/microbiología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...