Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 20(5): e1012189, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38713723

RESUMEN

Successful microbial colonization of the gastrointestinal (GI) tract hinges on an organism's ability to overcome the intense competition for nutrients in the gut between the host and the resident gut microbiome. Enteric pathogens can exploit ethanolamine (EA) in the gut to bypass nutrient competition. However, Klebsiella pneumoniae (K. pneumoniae) is an asymptomatic gut colonizer and, unlike well-studied enteric pathogens, harbors two genetically distinct ethanolamine utilization (eut) loci. Our investigation uncovered unique roles for each eut locus depending on EA utilization as a carbon or nitrogen source. Murine gut colonization studies demonstrated the necessity of both eut loci in the presence of intact gut microbiota for robust GI colonization by K. pneumoniae. Additionally, while some Escherichia coli gut isolates could metabolize EA, other commensals were incapable, suggesting that EA metabolism likely provides K. pneumoniae a selective advantage in gut colonization. Molecular and bioinformatic analyses unveiled the conservation of two eut loci among K. pneumoniae and a subset of the related taxa in the K. pneumoniae species complex, with the NtrC-RpoN regulatory cascade playing a pivotal role in regulation. These findings identify EA metabolism as a critical driver of K. pneumoniae niche establishment in the gut and propose microbial metabolism as a potential therapeutic avenue to combat K. pneumoniae infections.


Asunto(s)
Etanolamina , Microbioma Gastrointestinal , Infecciones por Klebsiella , Klebsiella pneumoniae , Klebsiella pneumoniae/metabolismo , Klebsiella pneumoniae/genética , Ratones , Animales , Etanolamina/metabolismo , Microbioma Gastrointestinal/fisiología , Infecciones por Klebsiella/microbiología , Infecciones por Klebsiella/metabolismo , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Ratones Endogámicos C57BL , Femenino
2.
Ann Surg ; 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38864230

RESUMEN

OBJECTIVE: To evaluate the persistence of intestinal microbiome dysbiosis and gut-plasma metabolomic perturbations following severe trauma or sepsis weeks after admission in patients experiencing chronic critical illness (CCI). SUMMARY: Trauma and sepsis can lead to gut dysbiosis and alterations in the plasma and fecal metabolome. However, the impact of these perturbations and correlations between gut dysbiosis and the plasma metabolome in chronic critical illness have not been studied. METHODS: A prospective observational cohort study was performed with healthy subjects, severe trauma patients, patients with sepsis residing in an intensive care unit (ICU) for 2-3 weeks. A high-throughput multi-omics approach was utilized to evaluate the gut microbial and gut-plasma metabolite responses in critically ill trauma and sepsis patients 14-21 days after ICU admission. RESULTS: Patients in the sepsis and trauma cohorts demonstrated strikingly depleted gut microbiome diversity, with significant alterations and specific pathobiome patterns in the microbiota composition compared to healthy subjects. Further subgroup analyses based on sex revealed resistance to changes in microbiome diversity among female trauma patients compared to healthy counterparts. Sex-specific changes in fecal metabolites were also observed after trauma and sepsis, while plasma metabolite changes were similar in both males and females. CONCLUSIONS: Dysbiosis induced by trauma and sepsis persists up to 14-21 days after onset and is sex-specific, underscoring the implication of pathobiome and entero-septic microbial-metabolite perturbations in post-sepsis and post-trauma CCI. This indicates resilience to infection or injury in females' microbiome and should inform and facilitate future precision/personalized medicine strategies in the intensive care unit.

3.
Microb Pathog ; 189: 106589, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38382627

RESUMEN

Comprehensive safety assessment of potential probiotic strains is crucial in the selection of risk-free strains for clinical translation. This study aimed to evaluate the biosafety of Limosilactobacillus fermentum NCDC 400, a potential probiotic strain, using oral toxicity tests in a Swiss albino mouse model. Mice were orally gavaged with low (108 CFU/mouse/day) and high (1010 CFU/mouse/day) doses of NCDC 400 for 14 (acute), 28 (subacute), and 90 (subchronic) days to assess behavioral, hematological, biochemical, immunological, and histological effects. The administration of NCDC 400 did not result in any observable adverse effects on general health parameters, including body weight, feed and water intake, and organ indices. Hematological and biochemical parameters, such as glucose, serum enzymes, urea, creatinine, serum minerals, total serum proteins, and lipid profile, remained largely unaffected by the test strain. Notably, NCDC 400 administration led to a significant reduction in harmful intestinal enzymes and improvement in gut health indices, as indicated by fecal pH, lactate, ammonia, and short-chain fatty acids. There were no instances of bacterial translocation of NCDC 400 to blood or extra-intestinal organs. Immune homeostasis was not adversely affected by repeated exposure to NCDC 400 in all three oral toxicity studies. Histopathological examination revealed no strain-related changes in various tissues. Based on these findings, a dose of 1010 CFU/mouse/day was considered as the No Observable Effect Level (NOEL) in healthy mice. In conclusion, this study demonstrates the safe and non-toxic behavior of L. fermentum NCDC 400. The results support and ensure the safety and suitability for clinical trials and eventual translation into clinical practice as potential probiotic.


Asunto(s)
Limosilactobacillus fermentum , Probióticos , Ratones , Animales , Modelos Animales de Enfermedad , Probióticos/metabolismo , Pruebas de Toxicidad
4.
Crit Care ; 28(1): 18, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212826

RESUMEN

BACKGROUND: Sepsis and trauma are known to disrupt gut bacterial microbiome communities, but the impacts and perturbations in the fungal (mycobiome) community after severe infection or injury, particularly in patients experiencing chronic critical illness (CCI), remain unstudied. METHODS: We assess persistence of the gut mycobiome perturbation (dysbiosis) in patients experiencing CCI following sepsis or trauma for up to two-to-three weeks after intensive care unit hospitalization. RESULTS: We show that the dysbiotic mycobiome arrays shift toward a pathobiome state, which is more susceptible to infection, in CCI patients compared to age-matched healthy subjects. The fungal community in CCI patients is largely dominated by Candida spp; while, the commensal fungal species are depleted. Additionally, these myco-pathobiome arrays correlate with alterations in micro-ecological niche involving specific gut bacteria and gut-blood metabolites. CONCLUSIONS: The findings reveal the persistence of mycobiome dysbiosis in both sepsis and trauma settings, even up to two weeks post-sepsis and trauma, highlighting the need to assess and address the increased risk of fungal infections in CCI patients.


Asunto(s)
Microbioma Gastrointestinal , Micobioma , Sepsis , Humanos , Disbiosis/complicaciones , Disbiosis/microbiología , Candida , Bacterias , Sepsis/complicaciones , Hongos
5.
Biochem Biophys Res Commun ; 682: 1-20, 2023 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-37788525

RESUMEN

Metabolic disorders are increasingly prevalent worldwide, leading to high rates of morbidity and mortality. The variety of metabolic illnesses can be addressed through personalized medicine. The goal of personalized medicine is to give doctors the ability to anticipate the best course of treatment for patients with metabolic problems. By analyzing a patient's metabolomic, proteomic, genetic profile, and clinical data, physicians can identify relevant diagnostic, and predictive biomarkers and develop treatment plans and therapy for acute and chronic metabolic diseases. To achieve this goal, real-time modeling of clinical data and multiple omics is essential to pinpoint underlying biological mechanisms, risk factors, and possibly useful data to promote early diagnosis and prevention of complex diseases. Incorporating cutting-edge technologies like artificial intelligence and machine learning is crucial for consolidating diverse forms of data, examining multiple variables, establishing databases of clinical indicators to aid decision-making, and formulating ethical protocols to address concerns. This review article aims to explore the potential of personalized medicine utilizing omics approaches for the treatment of metabolic disorders. It focuses on the recent advancements in genomics, epigenomics, proteomics, metabolomics, and nutrigenomics, emphasizing their role in revolutionizing personalized medicine.


Asunto(s)
Enfermedades Metabólicas , Proteómica , Humanos , Proteómica/métodos , Inteligencia Artificial , Genómica/métodos , Medicina de Precisión/métodos , Metabolómica/métodos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/terapia
6.
Clin Sci (Lond) ; 137(12): 963-978, 2023 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-37337946

RESUMEN

Sexual dimorphisms exist in multiple domains, from learning and memory to neurocognitive disease, and even in the immune system. Male sex has been associated with increased susceptibility to infection, as well as increased risk of adverse outcomes. Sepsis remains a major source of morbidity and mortality globally, and over half of septic patients admitted to intensive care are believed to suffer some degree of sepsis-associated encephalopathy (SAE). In the short term, SAE is associated with an increased risk of in-hospital mortality, and in the long term, has the potential for significant impairment of cognition, memory, and acceleration of neurocognitive disease. Despite increasing information regarding sexual dimorphism in neurologic and immunologic systems, research into these dimorphisms in sepsis-associated encephalopathy remains critically understudied. In this narrative review, we discuss how sex has been associated with brain morphology, chemistry, and disease, sexual dimorphism in immunity, and existing research into the effects of sex on SAE.


Asunto(s)
Encefalopatía Asociada a la Sepsis , Sepsis , Humanos , Masculino , Encefalopatía Asociada a la Sepsis/complicaciones , Caracteres Sexuales , Sepsis/complicaciones , Encéfalo
7.
Medicina (Kaunas) ; 59(9)2023 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-37763665

RESUMEN

The skeletal system is an extraordinary structure that serves multiple purposes within the body, including providing support, facilitating movement, and safeguarding vital organs. Moreover, it acts as a reservoir for essential minerals crucial for overall bodily function. The intricate interplay of bone cells plays a critical role in maintaining bone homeostasis, ensuring a delicate balance. However, various factors, both intrinsic and extrinsic, can disrupt this vital physiological process. These factors encompass genetics, aging, dietary and lifestyle choices, the gut microbiome, environmental toxins, and more. They can interfere with bone health through several mechanisms, such as hormonal imbalances, disruptions in bone turnover, direct toxicity to osteoblasts, increased osteoclast activity, immune system aging, impaired inflammatory responses, and disturbances in the gut-bone axis. As a consequence, these disturbances can give rise to a range of bone disorders. The regulation of bone's physiological functions involves an intricate network of continuous processes known as bone remodeling, which is influenced by various intrinsic and extrinsic factors within the organism. However, our understanding of the precise cellular and molecular mechanisms governing the complex interactions between environmental factors and the host elements that affect bone health is still in its nascent stages. In light of this, this comprehensive review aims to explore emerging evidence surrounding bone homeostasis, potential risk factors influencing it, and prospective therapeutic interventions for future management of bone-related disorders.


Asunto(s)
Densidad Ósea , Microbioma Gastrointestinal , Humanos , Osteoclastos , Envejecimiento , Homeostasis
8.
Toxicol Appl Pharmacol ; 447: 116066, 2022 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-35595072

RESUMEN

Carbon nanotubes (CNTs) are emerging environmental and occupational toxicants known to induce lung immunotoxicity. While the underlying mechanisms are evolving, it is yet unknown whether inhaled CNTs would cause abnormalities in gut microbiota (dysbiosis), and if such microbiota alteration plays a role in the modulation of CNT-induced lung immunotoxicity. It is also unknown whether co-exposure to tobacco smoke will modulate CNT effects. We compared the effects of lung exposure to multi-wall CNT, cigarette smoke extract (CSE), and their combination (CNT + CSE) in a 4-week chronic toxicity mouse model. The exposures induced differential perturbations in gut microbiome as evidenced by altered microbial α- and ß- diversity, indicating a lung-to-gut communication. The gut dysbiosis due to CNTs, unlike CSE, was characterized by an increase in Firmicutes/Bacteroidetes ratio typically associated with proinflammatory condition. Notably, while all three exposures reduced Proteobacteria, the CNT exposure and co-exposure induced appearance of Tenericutes and Cyanobacteria, respectively, implicating them as potential biomarkers of exposure. CNTs differentially induced certain lung proinflammatory mediators (TNF-α, IL-1ß, CCL2, CXCL5) whereas CNTs and CSE commonly induced other mediators (CXCL1 and TGF-ß). The co-exposure showed either a component-dominant effect or a summative effect for both dysbiosis and lung inflammation. Depletion of gut microbiota attenuated both the differentially-induced and commonly-induced (TGF-ß) lung inflammatory mediators as well as granulomas indicating gut-to-lung communication and a modulatory role of gut dysbiosis. Taken together, the results demonstrated gut dysbiosis as a systemic effect of inhaled CNTs and provided the first evidence of a bidirectional gut-lung crosstalk modulating CNT lung immunotoxicity.


Asunto(s)
Fumar Cigarrillos , Microbioma Gastrointestinal , Nanotubos de Carbono , Neumonía , Animales , Disbiosis/inducido químicamente , Disbiosis/complicaciones , Disbiosis/microbiología , Pulmón , Ratones , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente , Factor de Crecimiento Transformador beta
9.
Digestion ; 103(2): 133-140, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34749376

RESUMEN

BACKGROUND AND AIMS: Despite the reported salutary benefits of a Mediterranean diet (MD) on a wide variety of health conditions, the specific microbial changes associated with an MD within the gastrointestinal (GI) tract are not well studied. Specifically, although population and survey-based studies have shown microbial changes, there are no published data on how an MD alters the gut flora in a controlled setting. METHODS: We recruited 10 healthy subjects, each of whom gave a stool sample at baseline and then was provided with prepared meals of a "typical" American diet; after 2 weeks, a second stool sample was collected. All subjects were then provided with prepared meals based on the MD for another 2 weeks, followed by a final stool sample collection. Stool samples were batch analyzed with DNA extraction, and sequencing libraries were generated. Measures of bacterial diversity, species richness, and enterotypes were performed. RESULTS: All ten subjects tolerated the diets well. Bacterial diversity increased with an MD, as measured by alpha diversity via the Simpson index. Furthermore, there were significant differences in 5 bacterial genera between the 2 diets. CONCLUSION: This small pilot study of controlled diets demonstrates that the MD can rapidly alter the gut microbiome in healthy subjects at the level of global microbial diversity and individual genera. These data confirm the findings of previous observational studies and establish the feasibility of conducting longer term studies on the impact of the MD on the flora of the GI tract and its relationship to digestive diseases.


Asunto(s)
Dieta Mediterránea , Microbioma Gastrointestinal , Dieta , Heces/microbiología , Microbioma Gastrointestinal/genética , Voluntarios Sanos , Humanos , Proyectos Piloto
10.
Indian J Med Res ; 153(4): 492-502, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34380796

RESUMEN

BACKGROUND & OBJECTIVES: The vaginal microbiota undergoes subtle changes during pregnancy and may affect several aspects of pregnancy outcomes. There has been no comprehensive study characterizing the gestational vaginal and gut microbiota and the dynamics of the microbiota with oral probiotics among Indian women. Hence, the study was aimed to explore the microbiota of pregnant women with normal microbiota and bacterial vaginosis (BV) environments and the effect of oral probiotics on the microbiota and the BV status in these women. METHODS: Using high-throughput Illumina-MiSeq sequencing approach, the 16S rRNA gene amplicons were analyzed and the vaginal and gut microbiota of pregnant women with and without BV and pre- and post-probiotics (Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14) intervention for a month was characterized. RESULTS: The study revealed a compositional difference in the vaginal and gut microbiota between BV and healthy pregnant women. The vaginal microbiota of healthy women was characteristically predominated by Lactobacillus helveticus, followed by L. iners and L. gasseri; in contrast, women positive for BV harboured higher α-diversity and had lower abundance of L. helveticus. Similarly, Prevotella copri, a gut microbe, associated with normal environment was detected in the vaginal samples of all pregnant women without BV, it remained undetected in women with the infection, while all women with BV had Gardnerella vaginalis, which decreased significantly with probiotic treatment. Gut microbiota also revealed dominant abundance of P. copri in healthy women, whereas it was significantly lower in women with BV. The bacterial clade, P. copri abundance increased from 9.17 to 16.49 per cent in the probiotic group and reduced from 7.75 to 4.84 per cent in the placebo group. INTERPRETATION & CONCLUSIONS: This study showed gestational vaginal and gut microbiota differences in normal and BV environments. With probiotic treatment, the dynamics of L. helveticus and P. copri hint towards a possible role of probiotics in modulating the vaginal microbiota.


Asunto(s)
Microbioma Gastrointestinal , Probióticos , Vaginosis Bacteriana , Femenino , Humanos , Embarazo , Mujeres Embarazadas , Prevotella , ARN Ribosómico 16S/genética , Vagina
11.
Ann Nutr Metab ; 77 Suppl 2: 21-27, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33906194

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is the most common form of dementia, particularly in older adults, with clinical manifestations of progressive cognitive decline and functional impairment. The prevalence of AD and related dementia is mounting worldwide, but its etiology remains unresolved, with no available preventative or ameliorative therapy. Emerging evidence suggests that the gut microbiota of patients with AD is different from cognitively normal counterparts. SUMMARY: Communication between gut and brain (gut-brain axis) plays a crucial role in AD pathology. Bacteria inhabiting the gut strongly influence this gut-brain axis and thus may participate in AD pathology. Diet, one of the strongest modulators of gut microbiota, also strongly influences brain health and AD pathology. Gut microbiota metabolites including short-chain fatty acids, pro-inflammatory factors, and neurotransmitters may also affect AD pathogenesis and associated cognitive decline. Therefore, investigation of diet-microbiota-brain axis is important to better understand its contribution in AD pathology and its potential use as a target to prevent and treat AD. Herein, we discuss the link between AD and gut microbiota and ponder how microbiota modulation through nutritional approaches may offer avenues for discovering novel preventive and therapeutic strategies against AD. Key Message: A strong association exists between lifestyle factors and AD prevalence wherein unhealthy dietary factors have been linked to neurodegeneration. Specific prudent dietary patterns might help in preventing or delaying AD progression by affecting ß-amyloid production and tau processing and regulating AD-associated inflammation, metabolism and oxidative stress, plausibly via modulating gut microbiota.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Microbioma Gastrointestinal , Anciano , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Disfunción Cognitiva/etiología , Dieta , Microbioma Gastrointestinal/fisiología , Humanos
12.
Int J Mol Sci ; 22(4)2021 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-33562070

RESUMEN

The prevalence of type 2 diabetes mellitus (T2D) is increasing worldwide, and there are no long-term preventive strategies to stop this growth. Emerging research shows that perturbations in the gut microbiome significantly contribute to the development of T2D, while microbiome modulators may be beneficial for T2D prevention. However, microbiome modulators that are effective, safe, affordable, and able to be administered daily are not yet available. Based on our previous pro- and prebiotic studies, we developed a novel synbiotic yogurt comprised of human-origin probiotics and plant-based prebiotics and investigated its impact on diet- and streptozotocin-induced T2D in mice. We compared the effects of our synbiotic yogurt to those of a commercially available yogurt (control yogurt). Interestingly, we found that the feeding of the synbiotic yogurt significantly reduced the development of hyperglycemia (diabetes) in response to high-fat diet feeding and streptozotocin compared to milk-fed controls. Surprisingly, the control yogurt exacerbated diabetes progression. Synbiotic yogurt beneficially modulated the gut microbiota composition compared to milk, while the control yogurt negatively modulated it by significantly increasing the abundance of detrimental bacteria such as Proteobacteria and Enterobacteriaceae. In addition, the synbiotic yogurt protected pancreatic islet morphology compared to the milk control, while the control yogurt demonstrated worse effects on islets. These results suggest that our newly developed synbiotic yogurt protects against diabetes in mice and can be used as a therapeutic to prevent diabetes progression.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Diabetes Mellitus Tipo 2/prevención & control , Microbioma Gastrointestinal/fisiología , Simbióticos/administración & dosificación , Yogur/microbiología , Animales , Glucemia/análisis , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Tipo 2/dietoterapia , Dieta Alta en Grasa/efectos adversos , Humanos , Hiperglucemia/dietoterapia , Hiperglucemia/prevención & control , Intestinos/microbiología , Islotes Pancreáticos/fisiología , Lactobacillus/aislamiento & purificación , Masculino , Ratones , Ratones Endogámicos C57BL , Prebióticos/administración & dosificación , Probióticos/administración & dosificación
13.
Infect Immun ; 88(11)2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32839189

RESUMEN

An important yet poorly understood facet of the life cycle of a successful pathogen is host-to-host transmission. Hospital-acquired infections (HAI) resulting from the transmission of drug-resistant pathogens affect hundreds of millions of patients worldwide. Klebsiella pneumoniae, a Gram-negative bacterium, is notorious for causing HAI, with many of these infections difficult to treat, as K. pneumoniae has become multidrug resistant. Epidemiological studies suggest that K. pneumoniae host-to-host transmission requires close contact and generally occurs through the fecal-oral route. Here, we describe a murine model that can be utilized to study mucosal (oropharynx and gastrointestinal [GI]) colonization, shedding within feces, and transmission of K. pneumoniae through the fecal-oral route. Using an oral route of inoculation, and fecal shedding as a marker for GI colonization, we showed that K. pneumoniae can asymptomatically colonize the GI tract in immunocompetent mice and modifies the host GI microbiota. Colonization density within the GI tract and levels of shedding in the feces differed among the clinical isolates tested. A hypervirulent K. pneumoniae isolate was able to translocate from the GI tract and cause hepatic infection that mimicked the route of human infection. Expression of the capsule was required for colonization and, in turn, robust shedding. Furthermore, K. pneumoniae carrier mice were able to transmit to uninfected cohabitating mice. Lastly, treatment with antibiotics led to changes in the host microbiota and development of a transient supershedder phenotype, which enhanced transmission efficiency. Thus, this model can be used to determine the contribution of host and bacterial factors toward K. pneumoniae dissemination.


Asunto(s)
Enfermedades Gastrointestinales/microbiología , Infecciones por Klebsiella/transmisión , Animales , Modelos Animales de Enfermedad , Klebsiella pneumoniae , Ratones
14.
Int J Mol Sci ; 21(10)2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32414080

RESUMEN

The gut microbiome plays an important role in obesity and Type 2 diabetes (T2D); however, it remains unclear whether the gut microbiome could clarify the dietary versus genetic origin of these ailments. Moreover, studies examining the gut microbiome in diet- versus genetically induced obesity/T2D in the same experimental set-up are lacking. We herein characterized the gut microbiomes in three of the most widely used mouse models of obesity/T2D, i.e., genetically induced (leptin-deficient i.e., Lepob/ob; and leptin-receptor-deficient i.e., Lepdb/db) and high-fat diet (HFD)-induced obese (DIO)/T2D mice, with reference to their normal chow-fed (NC) and low-fat-diet-fed (LF) control counterparts. In terms of ß-diversity, Lepob/ob and Lepdb/db mice showed similarity to NC mice, whereas DIO and LF mice appeared as distinct clusters. The phylum- and genus-level compositions were relatively similar in NC, Lepob/ob, and Lepdb/db mice, whereas DIO and LF mice demonstrated distinct compositions. Further analyses revealed several unique bacterial taxa, metagenomic functional features, and their correlation patterns in these models. The data revealed that obesity/T2D driven by diet as opposed to genetics presents distinct gut microbiome signatures enriched with distinct functional capacities, and indicated that these signatures can distinguish diet- versus genetically induced obesity/T2D and, if extrapolated to humans, might offer translational potential in devising dietary and/or genetics-based therapies against these maladies.


Asunto(s)
Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal/genética , Leptina/genética , Obesidad/microbiología , Receptores de Leptina/genética , Animales , Diabetes Mellitus Tipo 2/genética , Dieta Alta en Grasa/efectos adversos , Humanos , Leptina/deficiencia , Masculino , Ratones , Ratones Endogámicos NOD/genética , Obesidad/genética , Obesidad/patología , Receptores de Leptina/deficiencia
15.
Ann Nutr Metab ; 73 Suppl 3: 4-11, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30041174

RESUMEN

Our gut microbiome plays a fundamental role in our health and disease. The microbial colonization of human gut begins immediately at birth and is an indispensable natural process that modulates our physiology and immunity. Recent studies are elegantly revealing how and when these microbes colonize the gut and what elements could potentially influence this natural phenomenon. The vertical mother-to-baby transmission of microbes is a crucial factor for normal development and maturation of newborn's immune, metabolic as well as neurological health. This important and delicate process of gut microbiota development may be impacted by various factors such as birth mode, type of feeding, gestational age at birth, antibiotics exposure in early life, surrounding environment and hygiene settings, and so on. Perturbations in early life gut microbial colonization have been associated with the development of several diseases such as diabetes, obesity, asthma, allergies, celiac disease, neurodevelopmental disorders, and so on. However, it remains unclear whether predisposition to these diseases is due to the lack of acquisition of the mother's (vaginal and perianal) microbes during birth or because of abnormal exposure to unsolicited bacteria. Hence, studies are required to scrutinize the colonization pattern of infant gut microbiome in context to birth mode and also to elucidate how long these differences could persist. In these contexts, we review and discuss some of the findings obtained from recent investigation of the gut microbiota composition in healthy Japanese infants and young adults born vaginally or by C-section.


Asunto(s)
Cesárea , Microbioma Gastrointestinal , Bacterias/clasificación , Parto Obstétrico , Femenino , Humanos , Recién Nacido , Japón , Embarazo , Vagina/microbiología , Adulto Joven
16.
Ann Nutr Metab ; 71 Suppl 1: 11-16, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28950279

RESUMEN

BACKGROUND: The intestinal epithelial layer is the chief barricade between the luminal contents and the host. A healthy homeostatic intestinal barrier is pivotal for maintaining gastrointestinal health, which impacts the overall health as it safeguards the gut-blood axis and checks gut microbes including potential pathogens from entering into the circulation. SUMMARY: Under healthy milieus, the intestinal barrier is generally very dynamic and effective, with luminal side being heavily infested with a wide variety of gut microbes while the basolateral side remains virtually sterile. However, certain conditions such as abnormal exposure to toxins, drugs, pathogens etc. or a state of hyper-inflammation due to disease conditions may weaken or destabilize the integrity of gut epithelia. A perturbed gut integrity and permeability ("leaky gut") may lead to microbial (bacterial) translocation, and the eventual leakage of bacteria or their metabolites into the circulation can make the host susceptible to various types of diseases via inducing chronic or acute inflammatory response. Key Message: Given a close association with gut integrity, bacterial translocation and inflammatory responses have recently emerged as a clinically important research field and have unveiled novel aspects of gut microbial ecology and various gastrointestinal, metabolic, and lifestyle diseases. This review aims to describe the significance of a healthy gut barrier integrity and permeability, as well as the factors and consequences associated with a compromised gut barrier, while discussing briefly the dietary approaches including probiotics and prebiotics that could ameliorate gut health by restoring gut environment and barrier integrity, thereby preventing bacterial translocation.


Asunto(s)
Traslocación Bacteriana/fisiología , Tracto Gastrointestinal/fisiología , Tracto Gastrointestinal/microbiología , Humanos
17.
BMC Microbiol ; 15: 219, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26482797

RESUMEN

BACKGROUND: Clostridium perfringens is a widespread pathogen, but the precise quantification of this subdominant gut microbe remains difficult due to its low fecal count (particularly in asymptomatic subjects) and also due to the presence of abundant polymerase-inhibitory substances in human feces. Also, information on the intestinal carriage of toxigenic C. perfringens strains in healthy subjects is sparse. Therefore, we developed a sensitive quantitative real-time PCR assays for quantification of C. perfringens in human feces by targeting its α-toxin and enterotoxin genes. To validate the assays, we finally observed the occurrence of α-toxigenic and enterotoxigenic C. perfringens in the fecal microbiota of healthy Japanese infants and young adults. METHODS: The plc-specific qPCR assay was newly validated, while primers for 16S rRNA and cpe genes were retrieved from literature. The assays were validated for specificity and sensitivity in pre-inoculated fecal samples, and were finally applied to quantify C. perfringens in stool samples from apparently healthy infants (n 124) and young adults (n 221). RESULTS: The qPCR assays were highly specific and sensitive, with a minimum detection limit of 10(3) bacterial cells/g feces. Alpha-toxigenic C. perfringens was detected in 36% infants and 33% adults, with counts ranging widely (10(3)-10(7) bacterial cells/g). Intriguingly, the mean count of α-toxigenic C. perfringens was significantly higher in infants (6.0±1.5 log10 bacterial cells/g), as compared to that in adults (4.8±1.2). Moreover, the prevalence of enterotoxigenic C. perfringens was also found to be significantly higher in infants, as compared to that in adults. The mean enterotoxigenic C. perfringens count was 5.9±1.9 and 4.8±0.8 log10 bacterial cells/g in infants and adults, respectively. CONCLUSIONS: These data indicate that some healthy infants and young adults carry α-toxigenic and enterotoxigenic C. perfringens at significant levels, and may be predisposed to related diseases. Thus, high fecal carriage of toxigenic C. perfringens in healthy children warrants further investigation on its potential sources and clinical significance in these subjects. In summary, we present a novel qPCR assay for sensitive and accurate quantification of α-toxigenic and enterotoxigenic C. perfringens in human feces, which should facilitate prospective studies of the gut microbiota.


Asunto(s)
Carga Bacteriana/métodos , Toxinas Bacterianas/genética , Proteínas de Unión al Calcio/genética , Infecciones por Clostridium/microbiología , Clostridium perfringens/aislamiento & purificación , Enterotoxinas/genética , Heces/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Fosfolipasas de Tipo C/genética , Adolescente , Adulto , Portador Sano/microbiología , Estudios de Cohortes , Femenino , Voluntarios Sanos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Sensibilidad y Especificidad , Adulto Joven
18.
Mediators Inflamm ; 2015: 793898, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26339142

RESUMEN

Oral cavity that harbors diverse bacterial populations could also act as a site of origin for spread of pathogenic microorganisms to different body sites, particularly in immunocompromised hosts, patients, the elderly, or the underprivileged. A number of recent publications have advocated that patients with periodontal diseases are more susceptible to metabolic endotoxemia, inflammation, obesity, type 2 diabetes, and other related systemic complications, concluding that periodontal diseases could be a potential contributing risk factor for a wide array of clinically important systemic diseases. However, despite a significant increase in the prevalence of periodontal infections and systemic diseases in the past few decades, the fundamental biological mechanisms of connection between these ailments are still not fully explicated. Consequently, the mechanisms by which this bidirectional damage occurs are being explored with a concentric vision to develop strategies that could prevent or control the complications of these ailments. This paper attempts to summarize and hypothesize the diverse mechanisms that hint to a certain connection between the two prevalent chronic situations.


Asunto(s)
Enfermedades Periodontales/complicaciones , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/etiología , Humanos , Obesidad/epidemiología , Obesidad/etiología , Enfermedades Periodontales/epidemiología , Factores de Riesgo
19.
Microbiome Res Rep ; 3(1): 9, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38455083

RESUMEN

Disturbances in the local and peripheral immune systems are closely linked to a wide range of diseases. In the context of neurodegenerative disorders such as Alzheimer's disease (AD), inflammation plays a crucial role, often appearing as a common manifestation despite the variability in the occurrence of other pathophysiological hallmarks. Thus, combating neuroinflammation holds promise in treating complex pathophysiological diseases like AD. Growing evidence suggests the gut microbiome's crucial role in shaping the pathogenesis of AD by influencing inflammatory mediators. Gut dysbiosis can potentially activate neuroinflammatory pathways through bidirectional signaling of the gut-brain axis; however, the precise mechanisms of this complex interweaved network remain largely unclear. In these milieus, this review attempts to summarize the contributing role of gut microbiome-mediated neuroinflammatory signals in AD pathophysiology, while also pondering potential mechanisms through which commensal and pathogenic gut microbes affect neuroinflammation. While certain taxa such as Roseburia and Escherichia have been strongly correlated with AD, other clades such as Bacteroides and Faecalibacterium exhibit variations at the species and strain levels. In order to disentangle the inflammatory aspects of neurodegeneration attributed to the gut microbiome, it is imperative that future mechanistic studies investigate the species/strain-level dependency of commensals, opportunistic, and pathogenic gut microbes that consistently show correlations with AD patients across multiple associative studies.

20.
Nutrients ; 16(12)2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38931317

RESUMEN

The consumption of almonds has been associated with several health benefits, particularly concerning cardiovascular and intestinal health. In this comprehensive review, we compile and deliberate studies investigating the effects of almond consumption on cardiovascular disease (CVD) risk factors and gut health. Almonds are rich in monounsaturated fats, fiber, vitamins, minerals, and polyphenols, which contribute to their health-promoting properties. Regular intake of almonds has been shown to improve lipid profiles by reducing LDL cholesterol and enhancing HDL functionality. Additionally, almonds aid in glycemic control, blood pressure reduction, and chronic inflammation amelioration, which are critical for cardiovascular health. The antioxidant properties of almonds, primarily due to their high vitamin E content, help in reducing oxidative stress markers. Furthermore, almonds positively influence body composition by reducing body fat percentage and central adiposity and enhancing satiety, thus aiding in weight management. Herein, we also contemplate the emerging concept of the gut-heart axis, where almond consumption appears to modulate the gut microbiome, promoting the growth of beneficial bacteria and increasing short-chain fatty acid production, particularly butyrate. These effects collectively contribute to the anti-inflammatory and cardioprotective benefits of almonds. By encompassing these diverse aspects, we eventually provide a systematic and updated perspective on the multifaceted benefits of almond consumption for cardiovascular health and gut microbiome, corroborating their broader consideration in dietary guidelines and public health recommendations for CVD risk reduction.


Asunto(s)
Enfermedades Cardiovasculares , Microbioma Gastrointestinal , Prunus dulcis , Humanos , Microbioma Gastrointestinal/fisiología , Enfermedades Cardiovasculares/prevención & control , Dieta , Antioxidantes , Factores de Riesgo de Enfermedad Cardiaca , Nueces
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA