Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43.741
Filtrar
1.
J Neuroinflammation ; 21(1): 120, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715051

RESUMEN

BACKGROUND: The human gut microbiome (GM) is involved in inflammation and immune response regulation. Dysbiosis, an imbalance in this ecosystem, facilitates pathogenic invasion, disrupts immune equilibrium, and potentially triggers diseases including various human leucocyte antigen (HLA)-B27-associated autoinflammatory and autoimmune diseases such as inflammatory bowel disease (IBD) and spondyloarthropathy (SpA). This study assesses compositional and functional alterations of the GM in patients with HLA-B27-associated non-infectious anterior uveitis (AU) compared to healthy controls. METHODS: The gut metagenomes of 20 patients with HLA-B27-associated non-infectious AU, 21 age- and sex-matched HLA-B27-negative controls, and 6 HLA-B27-positive healthy controls without a history of AU were sequenced using the Illumina NovaSeq 6000 platform for whole metagenome shotgun sequencing. To identify taxonomic and functional features with significantly different relative abundances between groups and to identify associations with clinical metadata, the multivariate association by linear models (MaAsLin) R package was applied. RESULTS: Significantly higher levels of the Eubacterium ramulus species were found in HLA-B27-negative controls (p = 0.0085, Mann-Whitney U-test). No significant differences in microbial composition were observed at all other taxonomic levels. Functionally, the lipid IVA biosynthesis pathway was upregulated in patients (p < 0.0001, Mann-Whitney U-test). A subgroup analysis comparing patients with an active non-infectious AU to their age- and sex-matched HLA-B27-negative controls, showed an increase of the species Phocaeicola vulgatus in active AU (p = 0.0530, Mann-Whitney U-test). An additional analysis comparing AU patients to age- and sex-matched HLA-B27-positive controls, showed an increase of the species Bacteroides caccae in controls (p = 0.0022, Mann-Whitney U-test). CONCLUSION: In our cohort, non-infectious AU development is associated with compositional and functional alterations of the GM. Further research is needed to assess the causality of these associations, offering potentially novel therapeutic strategies.


Asunto(s)
Microbioma Gastrointestinal , Antígeno HLA-B27 , Uveítis Anterior , Humanos , Antígeno HLA-B27/genética , Antígeno HLA-B27/inmunología , Femenino , Masculino , Microbioma Gastrointestinal/fisiología , Persona de Mediana Edad , Uveítis Anterior/microbiología , Uveítis Anterior/inmunología , Adulto , Estudios de Casos y Controles , Anciano
2.
Microbiome ; 12(1): 80, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715137

RESUMEN

BACKGROUND: Antibiotic exposure can occur in medical settings and from environmental sources. Long-term effects of brief antibiotic exposure in early life are largely unknown. RESULTS: Post a short-term treatment by ceftriaxone to C57BL/6 mice in early life, a 14-month observation was performed using 16S rRNA gene-sequencing technique, metabolomics analysis, and metagenomics analysis on the effects of ceftriaxone exposure. Firstly, the results showed that antibiotic pre-treatment significantly disturbed gut microbial α and ß diversities (P < 0.05). Both Chao1 indices and Shannon indices manifested recovery trends over time, but they didn't entirely recover to the baseline of control throughout the experiment. Secondly, antibiotic pre-treatment reduced the complexity of gut molecular ecological networks (MENs). Various network parameters were affected and manifested recovery trends over time with different degrees, such as nodes (P < 0.001, R2 = 0.6563), links (P < 0.01, R2 = 0.4543), number of modules (P = 0.0672, R2 = 0.2523), relative modularity (P = 0.6714, R2 = 0.0155), number of keystones (P = 0.1003, R2 = 0.2090), robustness_random (P = 0.79, R2 = 0.0063), and vulnerability (P = 0.0528, R2 = 0.28). The network parameters didn't entirely recover. Antibiotic exposure obviously reduced the number of key species in gut MENs. Interestingly, new keystones appeared during the recovery process of network complexity. Changes in network stability might be caused by variations in network complexity, which supports the ecological theory that complexity begets stability. Besides, the metabolism profiles of the antibiotic group and control were significantly different. Correlation analysis showed that antibiotic-induced differences in gut microbial metabolism were related to MEN changes. Antibiotic exposure also caused long-term effects on gut microbial functional networks in mice. CONCLUSIONS: These results suggest that short-term antibiotic exposure in early life will cause long-term negative impacts on gut microbial diversity, MENs, and microbial metabolism. Therefore, great concern should be raised about children's brief exposure to antibiotics if the results observed in mice are applicable to humans. Video Abstract.


Asunto(s)
Antibacterianos , Bacterias , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , ARN Ribosómico 16S , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Antibacterianos/farmacología , Antibacterianos/efectos adversos , Ratones , ARN Ribosómico 16S/genética , Bacterias/genética , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Ceftriaxona/farmacología , Metagenómica/métodos , Masculino , Metabolómica , Heces/microbiología
3.
Trials ; 25(1): 307, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715143

RESUMEN

BACKGROUND: Aging has been associated with a progressive loss of skeletal muscle quality, quantity and strength, which may result in a condition known as sarcopenia, leading to a decline in physical performance, loss of independence and reduced quality of life. While the cause of impaired physical functioning observed in elderly populations appears to be multifactorial, recent evidence suggests that age-associated alterations in gut microbiota could be a contributing factor. The primary objective will be to assess the effects of a dietary synbiotic formulation on sarcopenia-related functional outcomes such as handgrip strength, gait speed and physical performance within older individuals living independently. The secondary objective will be to examine associations between changes in gut microbiota composition, functional performance and lean muscle mass. METHODS: Seventy-four elderly (60-85 years) participants will be randomized in a double-blind, placebo-controlled fashion to either an intervention or control group. The intervention group (n = 37) will receive oral synbiotic formulation daily for 16 weeks. The control group (n = 37) will receive placebo. Assessments of physical performance (including Short Physical Performance Battery, handgrip strength and timed up-and-go tests) and muscle ultrasonography will be performed at 4 time points (baseline and weeks 8, 16 and 20). Likewise, body composition via bioelectric impedance analysis and blood and stool samples will be collected at each time point. Dual-energy X-ray absorptiometry will be performed at baseline and week 16. The primary outcomes will be between-group changes in physical performance from baseline to 16 weeks. Secondary outcomes include changes in body composition, muscle mass and architecture, fecal microbiota composition and diversity, and fecal and plasma metabolomics. DISCUSSION: Gut-modulating supplements appear to be effective in modifying gut microbiota composition in healthy older adults. However, it is unclear whether these changes translate into functional and/or health improvements. In the present study, we will investigate the effects of a synbiotic formulation on measures of physical performance, strength and muscle health in healthy older populations. TRIAL REGISTRATION: This study was prospectively registered with the Australian New Zealand Clinical Trials Registry (ACTRN12622000652774) in May 2022.


Asunto(s)
Microbioma Gastrointestinal , Fuerza de la Mano , Fuerza Muscular , Músculo Esquelético , Ensayos Clínicos Controlados Aleatorios como Asunto , Sarcopenia , Simbióticos , Humanos , Método Doble Ciego , Anciano , Simbióticos/administración & dosificación , Anciano de 80 o más Años , Sarcopenia/fisiopatología , Sarcopenia/prevención & control , Masculino , Persona de Mediana Edad , Femenino , Australia , Rendimiento Físico Funcional , Suplementos Dietéticos , Composición Corporal , Resultado del Tratamiento , Velocidad al Caminar , Pueblos de Australasia
4.
Acta Biochim Pol ; 71: 12020, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38721310

RESUMEN

Objective: To explore the difference in intestinal microecology between patients with preeclampsia and pregnant women at different stages of pregnancy. Methods: From January 2020 to January 2022, clinical data, including blood routine, lipid profile, and renal function indicators, were gathered from a cohort consisting of 5 cases of preeclampsia and 34 cases of non-preeclampsia. The non-preeclampsia group was further categorized into 6 cases in the First trimester, 13 cases in the Second trimester, and 15 cases in the Third trimester. The data collection took place at the Obstetrics Department of the Maternal and Child Health Hospital of Hubei Province. Additionally, fecal samples were obtained from each subject for 16S rDNA gene sequencing and subsequent analysis. The clinical data and composition characteristics of the gut microbiota in each group were analyzed, and the correlation between gut microbiota and clinical data was analyzed by the Spearman correlation analysis method. Results: In comparison to pregnant women without preeclampsia, preeclampsia patients exhibited a statistically significant elevation in blood routine parameters (WBC, N, L, and PLT count), a rise in lipid-related indicators (TC, TG, and LDL-C levels), a reduction in HDL-C levels, and an increase in renal function-related indicators (Cr, BUN, UA and Pro levels). Compared with non-preeclampsia pregnant women, preeclampsia women exhibited an augmented diversity of gut microbiota. Differences in gut microbiota composition between the two groups were observed at the gate and genus levels. Moreover, there are significant differences in the composition of gut microbiota between the preeclampsia group and the third-trimester group in terms of genus and species, and this difference is mainly caused by Prevotella and s_ Bacteroides_ Uniformis and Ruminococcus_ bromii. In addition, actinobacteria, bifidobacterium at the genus level, and Ruminococcus_bromii at the species level are positively correlated with clinically relevant indicators (excluding HDL-C). Conclusion: There are significant differences in gut microbiota between preeclampsia pregnant women and late pregnancy pregnant without preeclampsia, including Prevotella and Bacteroides_ Uniformis, and Ruminococcus_ bromii. In addition, these differential bacteria are correlated with most clinical indicators. However, additional comprehensive analysis is required to ascertain the functional correlation between these bacteria and clinical indicators.


Asunto(s)
Microbioma Gastrointestinal , Preeclampsia , Humanos , Embarazo , Preeclampsia/microbiología , Femenino , Adulto , Heces/microbiología , ARN Ribosómico 16S/genética
5.
Am J Reprod Immunol ; 91(5): e13859, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38722063

RESUMEN

Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.


Asunto(s)
Aborto Habitual , Humanos , Femenino , Embarazo , Aborto Habitual/inmunología , Tolerancia Inmunológica , Intercambio Materno-Fetal/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoterapia/métodos
6.
Cell Host Microbe ; 32(5): 623-624, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723597

RESUMEN

Common nutrients in our diet often affect our health through unexpected mechanisms. In a recent issue of Nature, Scott et al. show gut microbes convert dietary tryptophan into metabolites activating intestinal dopamine receptors, which can block attachment of bacterial pathogens to host cells.


Asunto(s)
Dopamina , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Dopamina/metabolismo , Humanos , Receptores Dopaminérgicos/metabolismo , Animales , Triptófano/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Bacterias/metabolismo , Interacciones Huésped-Patógeno , Adhesión Bacteriana
7.
Cell Host Microbe ; 32(5): 625-626, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723598

RESUMEN

Fungi colonize the mammalian gastrointestinal (GI) tract and can adopt both commensal and opportunistic lifestyles. In a recent issue of Nature, Liang et al. unraveled the complex interplay between Candida morphotypes and the gut bacterial microbiota and described a key role for candidalysin in gut colonization.1.


Asunto(s)
Candida , Microbioma Gastrointestinal , Tracto Gastrointestinal , Simbiosis , Microbioma Gastrointestinal/fisiología , Humanos , Tracto Gastrointestinal/microbiología , Animales , Candida/fisiología , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética
8.
Cell Host Microbe ; 32(5): 630-632, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723600

RESUMEN

The gut microbiota has the capacity to metabolize food-derived molecules. In this issue of Cell Host & Microbe, Li et al. explore how some bacterial species of the gut microbiota can deplete amino acids in the gut lumen, modulating the amino acid landscape and energy metabolism of the host.


Asunto(s)
Aminoácidos , Metabolismo Energético , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Aminoácidos/metabolismo , Humanos , Bacterias/metabolismo , Bacterias/genética , Animales , Interacciones Microbiota-Huesped , Tracto Gastrointestinal/microbiología
9.
Cell Host Microbe ; 32(5): 637-638, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723603

RESUMEN

Gut bacteria are thought to contribute to neurodevelopmental disorders, but whether they are causal or predictive of disease remains unclear. In a prospective longitudinal study of thousands of children, Ahrens et al. generate evidence for the role of the gut microbiome in neurodevelopmental disorders while highlighting important open questions.


Asunto(s)
Microbioma Gastrointestinal , Trastornos del Neurodesarrollo , Humanos , Niño , Estudios Longitudinales , Bacterias/genética , Estudios Prospectivos
10.
Cell Host Microbe ; 32(5): 639-650, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38723604

RESUMEN

There is rapidly growing awareness of microbiome assembly and function in early-life gut health. Although many factors, such as antibiotic use and highly processed diets, impinge on this process, most research has focused on people residing in high-income countries. However, much of the world's population lives in low- and middle-income countries (LMICs), where, in addition to erratic antibiotic use and suboptimal diets, these groups experience unique challenges. Indeed, many children in LMICs are infected with intestinal helminths. Although helminth infections are strongly associated with diverse developmental co-morbidities and induce profound microbiome changes, few studies have directly examined whether intersecting pathways between these components of the holobiont shape health outcomes in early life. Here, we summarize microbial colonization within the first years of human life, how helminth-mediated changes to the gut microbiome may affect postnatal growth, and why more research on this relationship may improve health across the lifespan.


Asunto(s)
Microbioma Gastrointestinal , Helmintiasis , Helmintos , Microbioma Gastrointestinal/fisiología , Humanos , Helmintos/fisiología , Animales , Lactante , Parasitosis Intestinales
12.
Clin Exp Rheumatol ; 42(5): 1097-1103, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38743445

RESUMEN

The relationship between intestinal microbiota and arthritis has garnered significant attention, with emerging evidence suggesting a potential association between dysbiosis and various forms of inflammatory arthropathies. While observational studies have provided valuable insights into microbiota alterations in patients with arthritis, establishing causality remains challenging. Observational data, influenced by multiple confounders such as environmental factors, medication effects, and dietary habits, are insufficient to conclusively determine whether microbiota changes are somehow causally linked to arthritis. The heterogeneity of results across independent studies further complicates interpretation. To further support this hypothesis, interventional randomised trials are deemed necessary, yet their implementation in this area presents significant technical limitations. Experimental animal models offer insights into potential pathogenic mechanisms linking dysbiosis to arthritis, including compromised intestinal barrier function, the role of microbiota-derived metabolites and molecular mimicry. However, conflicting findings underscore the complexity of hostmicrobiota interactions and the challenges in establishing causality.Efforts to modulate the microbiota for arthritis treatment or prevention have shown promise, yet efficacy and applicability remains uncertain. Antibacterial drugs, dietary interventions, probiotics, and faecal microbiota transplantation have been explored, but their clinical utility awaits further validation. In conclusion, while the association between intestinal microbiota and arthritis is increasingly recognised, establishing causality remains elusive.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Humanos , Animales , Probióticos/uso terapéutico , Artritis/microbiología , Trasplante de Microbiota Fecal , Interacciones Huésped-Patógeno , Factores de Riesgo
13.
Medicine (Baltimore) ; 103(19): e38088, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728472

RESUMEN

Microbiota modulation, the intentional change in the structure and function of the microbial community, is an emerging trajectory that holds the promise to mitigate an infinite number of health issues. The present review illustrates the underlying principles of microbiota modulation and the various applications of this fundamental process to human health, healthcare management, and pharmacologic interventions. Different strategies, directing on dietary interventions, fecal microbiota transplantation, treatment with antibiotics, bacteriophages, microbiome engineering, and modulation of the immune system, are described in detail. This therapeutic implication is reflected in clinical applications to gastrointestinal disorders and immune-mediated diseases for microbiota-modulating agents. In addition to this, the review outlines the challenges of translating researched outcomes into clinical practice to consider safety and provides insights into future research directions of this rapidly developing area.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Humanos , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal/fisiología , Antibacterianos/uso terapéutico , Probióticos/uso terapéutico , Enfermedades Gastrointestinales/terapia , Enfermedades Gastrointestinales/microbiología
14.
Molecules ; 29(9)2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38731645

RESUMEN

Ulcerative colitis (UC), as a chronic inflammatory disease, presents a global public health threat. However, the mechanism of Poria cocos (PC) in treating UC remains unclear. Here, LC-MS/MS was carried out to identify the components of PC. The protective effect of PC against UC was evaluated by disease activity index (DAI), colon length and histological analysis in dextran sulfate sodium (DSS)-induced UC mice. ELISA, qPCR, and Western blot tests were conducted to assess the inflammatory state. Western blotting and immunohistochemistry techniques were employed to evaluate the expression of tight junction proteins. The sequencing of 16S rRNA was utilized for the analysis of gut microbiota regulation. The results showed that a total of fifty-two nutrients and active components were identified in PC. After treatment, PC significantly alleviated UC-associated symptoms including body weight loss, shortened colon, an increase in DAI score, histopathologic lesions. PC also reduced the levels of inflammatory cytokines TNF-α, IL-6, and IL-1ß, as evidenced by the suppressed NF-κB pathway, restored the tight junction proteins ZO-1 and Claudin-1 in the colon, and promoted the diversity and abundance of beneficial gut microbiota. Collectively, these findings suggest that PC ameliorates colitis symptoms through the reduction in NF-κB signaling activation to mitigate inflammatory damage, thus repairing the intestinal barrier, and regulating the gut microbiota.


Asunto(s)
Colitis Ulcerosa , Sulfato de Dextran , Microbioma Gastrointestinal , FN-kappa B , Transducción de Señal , Wolfiporia , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , FN-kappa B/metabolismo , Ratones , Transducción de Señal/efectos de los fármacos , Wolfiporia/química , Masculino , Modelos Animales de Enfermedad , Citocinas/metabolismo , Colon/patología , Colon/metabolismo , Colon/efectos de los fármacos , Colon/microbiología , Proteínas de Uniones Estrechas/metabolismo , Ratones Endogámicos C57BL
15.
Int J Mol Sci ; 25(9)2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38732038

RESUMEN

The gut microbiota and short chain fatty acids (SCFA) have been associated with immune regulation and autoimmune diseases. Autoimmune kidney diseases arise from a loss of tolerance to antigens, often with unclear triggers. In this review, we explore the role of the gut microbiome and how disease, diet, and therapy can alter the gut microbiota consortium. Perturbations in the gut microbiota may systemically induce the translocation of microbiota-derived inflammatory molecules such as liposaccharide (LPS) and other toxins by penetrating the gut epithelial barrier. Once in the blood stream, these pro-inflammatory mediators activate immune cells, which release pro-inflammatory molecules, many of which are antigens in autoimmune diseases. The ratio of gut bacteria Bacteroidetes/Firmicutes is associated with worse outcomes in multiple autoimmune kidney diseases including lupus nephritis, MPO-ANCA vasculitis, and Goodpasture's syndrome. Therapies that enhance SCFA-producing bacteria in the gut have powerful therapeutic potential. Dietary fiber is fermented by gut bacteria which in turn release SCFAs that protect the gut barrier, as well as modulating immune responses towards a tolerogenic anti-inflammatory state. Herein, we describe where the current field of research is and the strategies to harness the gut microbiome as potential therapy.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/inmunología , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Animales , Ácidos Grasos Volátiles/metabolismo , Enfermedades Renales/microbiología , Enfermedades Renales/inmunología , Enfermedades Renales/terapia
16.
Parasit Vectors ; 17(1): 217, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734668

RESUMEN

BACKGROUND: Gut bacteria, which serve as essential modulators, exert a significant impact on insect physiology and behavior and have substantial application potential in pest management. The dynamics of gut bacteria and their impact on Phortica okadai behavior remain unclear. METHODS: In this study, the dynamics of gut bacteria at different developmental stages in P. okadai were analyzed using 16S ribosomal RNA (rRNA) gene sequencing, and the species and abundance of gut bacteria that affect host behavior were examined via behavioral experiments. RESULTS: A total of 19 phyla, 29 classes, 74 orders, 101 species, and 169 genera were identified. The results of the behavioral experiments indicated that the species Lactiplantibacillus argentoratensis, Acetobacter tropicalis, Leuconostoc citreum, and Levilactobacillus brevis effectively influenced the feeding preference of P. okadai, and the single-bacterium-seeded P. okadai exhibited feeding preferences distinct from those of the germ-free (GF) and wild-type P. okadai. CONCLUSIONS: The species and relative abundance of gut bacteria together positively impact P. okadai behavior. Lactiplantibacillus argentoratensis, as the most attractive bacteria to P. okadai, presents opportunities for novel pest control strategies targeting this vector and agricultural pest.


Asunto(s)
Bacterias , Microbioma Gastrointestinal , ARN Ribosómico 16S , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , ARN Ribosómico 16S/genética , Conducta Animal , Conducta Alimentaria , Tephritidae/microbiología , Tephritidae/fisiología
17.
Sci Rep ; 14(1): 10814, 2024 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734695

RESUMEN

Chikungunya virus (CHIKV) poses a significant global health threat, re-emerging as a mosquito-transmitted pathogen that caused high fever, rash, and severe arthralgia. In Thailand, a notable CHIKV outbreak in 2019-2020 affected approximately 20,000 cases across 60 provinces, underscoring the need for effective mosquito control protocols. Previous studies have highlighted the role of midgut bacteria in the interaction between mosquito vectors and pathogen infections, demonstrating their ability to protect the insect from invading pathogens. However, research on the midgut bacteria of Aedes (Ae.) aegypti, the primary vector for CHIKV in Thailand remains limited. This study aims to characterize the bacterial communities in laboratory strains of Ae. aegypti, both infected and non-infected with CHIKV. Female mosquitoes from a laboratory strain of Ae. aegypti were exposed to a CHIKV-infected blood meal through membrane feeding, while the control group received a non-infected blood meal. At 7 days post-infection (dpi), mosquito midguts were dissected for 16S rRNA gene sequencing to identify midgut bacteria, and CHIKV presence was confirmed by E1-nested RT-PCR using mosquito carcasses. The study aimed to compare the bacterial communities between CHIKV-infected and non-infected groups. The analysis included 12 midgut bacterial samples, divided into three groups: CHIKV-infected (exposed and infected), non-infected (exposed but not infected), and non-exposed (negative control). Alpha diversity indices and Bray-Curtis dissimilarity matrix revealed significant differences in bacterial profiles among the three groups. The infected group exhibited an increased abundance of bacteria genus Gluconobacter, while Asaia was prevalent in both non-infected and negative control groups. Chryseobacterium was prominent in the negative control group. These findings highlight potential alterations in the distribution and abundance of gut microbiomes in response to CHIKV infection status. This study provides valuable insights into the dynamic relationship between midgut bacteria and CHIKV, underscoring the potential for alterations in bacterial composition depending on infection status. Understanding the relationships between mosquitoes and their microbiota holds promise for developing new methods and tools to enhance existing strategies for disease prevention and control. This research advances our understanding of the circulating bacterial composition, opening possibilities for new approaches in combating mosquito-borne diseases.


Asunto(s)
Aedes , Virus Chikungunya , Microbioma Gastrointestinal , Mosquitos Vectores , Animales , Femenino , Aedes/microbiología , Aedes/virología , Bacterias/genética , Bacterias/clasificación , Bacterias/aislamiento & purificación , Fiebre Chikungunya/transmisión , Fiebre Chikungunya/virología , Virus Chikungunya/genética , Virus Chikungunya/aislamiento & purificación , Virus Chikungunya/fisiología , Mosquitos Vectores/microbiología , Mosquitos Vectores/virología , ARN Ribosómico 16S/genética , Tailandia
18.
Front Cell Infect Microbiol ; 14: 1387126, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38736752

RESUMEN

Introduction: We examined the gut microbiota of travellers returning from tropical areas with and without traveller's diarrhoea (TD) and its association with faecal lipocalin-2 (LCN2) levels. Methods: Participants were recruited at the Hospital Clinic of Barcelona, Spain, and a single stool sample was collected from each individual to perform the diagnostic of the etiological agent causing gastrointestinal symptoms as well as to measure levels of faecal LCN2 as a biomarker of gut inflammation. We also characterised the composition of the gut microbiota by sequencing the region V3-V4 from the 16S rRNA gene, and assessed its relation with the clinical presentation of TD and LCN2 levels using a combination of conventional statistical tests and unsupervised machine learning approaches. Results: Among 61 participants, 45 had TD, with 40% having identifiable etiological agents. Surprisingly, LCN2 levels were similar across groups, suggesting gut inflammation occurs without clinical TD symptoms. Differential abundance (DA) testing highlighted a microbial profile tied to high LCN2 levels, marked by increased Proteobacteria and Escherichia-Shigella, and decreased Firmicutes, notably Oscillospiraceae. UMAP analysis confirmed this profile's association, revealing distinct clusters based on LCN2 levels. The study underscores the discriminatory power of UMAP in capturing meaningful microbial patterns related to clinical variables. No relevant differences in the gut microbiota composition were found between travellers with or without TD. Discussion: The findings suggest a correlation between gut microbiome and LCN2 levels during travel, emphasising the need for further research to discern the nature of this relationship.


Asunto(s)
Diarrea , Heces , Microbioma Gastrointestinal , Lipocalina 2 , ARN Ribosómico 16S , Humanos , Lipocalina 2/metabolismo , Heces/microbiología , Heces/química , Masculino , Adulto , Femenino , ARN Ribosómico 16S/genética , Persona de Mediana Edad , Diarrea/microbiología , España , Viaje , Biomarcadores , Inflamación/microbiología , Adulto Joven , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación
19.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719750

RESUMEN

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Asunto(s)
Candida albicans , Enfermedad Celíaca , Homeostasis , Mastocitos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/microbiología , Enfermedad Celíaca/metabolismo , Humanos , Candida albicans/patogenicidad , Candida albicans/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Microbioma Gastrointestinal/inmunología , Disbiosis/inmunología , Candidiasis/inmunología , Candidiasis/microbiología , Animales , Candida/patogenicidad , Candida/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
20.
Nat Commun ; 15(1): 3872, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719797

RESUMEN

The gut microbiota and microglia play critical roles in Alzheimer's disease (AD), and elevated Bacteroides is correlated with cerebrospinal fluid amyloid-ß (Aß) and tau levels in AD. We hypothesize that Bacteroides contributes to AD by modulating microglia. Here we show that administering Bacteroides fragilis to APP/PS1-21 mice increases Aß plaques in females, modulates cortical amyloid processing gene expression, and down regulates phagocytosis and protein degradation microglial gene expression. We further show that administering Bacteroides fragilis to aged wild-type male and female mice suppresses microglial uptake of Aß1-42 injected into the hippocampus. Depleting murine Bacteroidota with metronidazole decreases amyloid load in aged 5xFAD mice, and activates microglial pathways related to phagocytosis, cytokine signaling, and lysosomal degradation. Taken together, our study demonstrates that members of the Bacteroidota phylum contribute to AD pathogenesis by suppressing microglia phagocytic function, which leads to impaired Aß clearance and accumulation of amyloid plaques.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Ratones Transgénicos , Microglía , Fagocitosis , Placa Amiloide , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/microbiología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Placa Amiloide/metabolismo , Femenino , Ratones , Masculino , Bacteroides fragilis/metabolismo , Microbioma Gastrointestinal , Humanos , Ratones Endogámicos C57BL , Hipocampo/metabolismo , Hipocampo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA