Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.065
Filter
1.
J Transl Med ; 22(1): 773, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152439

ABSTRACT

BACKGROUND: The prevalence of chronic kidney disease (CKD) is on the rise, posing a significant public health challenge. Although gut microbiome dysbiosis has been implicated in the impairment of kidney functions, the existence of pathological subtypes-linked differences remains largely unknown. We aimed to characterize the intestinal microbiota in patients with membranous nephropathy (MN), IgA nephropathy (IgAN), minimal change disease (MCD), and ischemic renal injury (IRI) in order to investigate the intricate relationship between intestinal microbiota and CKD across different subtypes. METHODS: We conducted a cross-sectional study involving 94 patients with various pathological patterns of CKD and 54 healthy controls (HCs). The clinical parameters were collected, and stool samples were obtained from each participant. Gut microbial features were analyzed using 16S rRNA sequencing and taxon annotation to compare the HC, CKD, MN, IgAN, MCD, and IRI groups. RESULTS: The CKD subjects exhibited significantly reduced alpha diversity, modified community structures, and disrupted microbial composition and potential functions compared to the control group. The opportunistic pathogen Klebsiella exhibited a significant enrichment in patients with CKD, whereas Akkermansia showed higher abundance in HCs. The study further revealed the presence of heterogeneity in intestinal microbial signatures across diverse CKD pathological types, including MN, IgAN, MCD, and IRI. The depression of the family Lachnospiraceae and the genus Bilophila was prominently observed exclusively in patients with MN, while suppressed Streptococcus was detected only in individuals with MCD, and a remarkable expansion of the genus Escherichia was uniquely found in cases of IRI. The study also encompassed the development of classifiers employing gut microbial diagnostic markers to accurately discriminate between distinct subtypes of CKD. CONCLUSIONS: The dysregulation of gut microbiome was strongly correlated with CKD, exhibiting further specificity towards distinct pathological patterns. Our study emphasizes the significance of considering disease subtypes when assessing the impact of intestinal microbiota on the development, diagnosis, and treatment of CKD.


Subject(s)
Gastrointestinal Microbiome , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/pathology , Male , Female , Middle Aged , Case-Control Studies , Adult , RNA, Ribosomal, 16S/genetics , Cross-Sectional Studies , Dysbiosis/microbiology , Dysbiosis/complications , Feces/microbiology
2.
J Transl Med ; 22(1): 729, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103909

ABSTRACT

BACKGROUND: Metabolic syndrome (MetS) is highly prevalent in individuals with schizophrenia (SZ), leading to negative consequences like premature mortality. Gut dysbiosis, which refers to an imbalance of the microbiota, and chronic inflammation are associated with both SZ and MetS. However, the relationship between gut dysbiosis, host immunological dysfunction, and SZ comorbid with MetS (SZ-MetS) remains unclear. This study aims to explore alterations in gut microbiota and their correlation with immune dysfunction in SZ-MetS, offering new insights into its pathogenesis. METHODS AND RESULTS: We enrolled 114 Chinese patients with SZ-MetS and 111 age-matched healthy controls from Zhejiang, China, to investigate fecal microbiota using Illumina MiSeq sequencing targeting 16 S rRNA gene V3-V4 hypervariable regions. Host immune responses were assessed using the Bio-Plex Pro Human Cytokine 27-Plex Assay to examine cytokine profiles. In SZ-MetS, we observed decreased bacterial α-diversity and significant differences in ß-diversity. LEfSe analysis identified enriched acetate-producing genera (Megamonas and Lactobacillus), and decreased butyrate-producing bacteria (Subdoligranulum, and Faecalibacterium) in SZ-MetS. These altered genera correlated with body mass index, the severity of symptoms (as measured by the Scale for Assessment of Positive Symptoms and Scale for Assessment of Negative Symptoms), and triglyceride levels. Altered bacterial metabolic pathways related to lipopolysaccharide biosynthesis, lipid metabolism, and various amino acid metabolism were also found. Additionally, SZ-MetS exhibited immunological dysfunction with increased pro-inflammatory cytokines, which correlated with the differential genera. CONCLUSION: These findings suggested that gut microbiota dysbiosis and immune dysfunction play a vital role in SZ-MetS development, highlighting potential therapeutic approaches targeting the gut microbiota. While these therapies show promise, further mechanistic studies are needed to fully understand their efficacy and safety before clinical implementation.


Subject(s)
Gastrointestinal Microbiome , Metabolic Syndrome , Schizophrenia , Adult , Female , Humans , Male , Middle Aged , Case-Control Studies , China , Comorbidity , Cytokines/metabolism , Dysbiosis/microbiology , Dysbiosis/immunology , Dysbiosis/complications , East Asian People , Feces/microbiology , Immunity , Metabolic Syndrome/microbiology , Metabolic Syndrome/immunology , Metabolic Syndrome/complications , Schizophrenia/microbiology , Schizophrenia/immunology , Schizophrenia/complications
3.
Nutrients ; 16(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39125448

ABSTRACT

It is now established that patients with rheumatoid arthritis (RA) have an increased risk of developing cervical cancer (CC) or its precursor, cervical intraepithelial neoplasia (CIN). However, the underlying mechanisms of this association have not been elucidated. RA is characterized by unresolved chronic inflammation. It is suggested that human papillomavirus (HPV) infection in RA patients exacerbates inflammation, increasing the risk of CC. The tumor microenvironment in RA patients with CC is also marked by chronic inflammation, which aggravates the manifestations of both conditions. Gut and vaginal dysbiosis are also considered potential mechanisms that contribute to the chronic inflammation and aggravation of RA and CC manifestations. Numerous clinical and pre-clinical studies have demonstrated the beneficial effects of various nutritional approaches to attenuate chronic inflammation, including polyunsaturated fatty acids and their derivatives, specialized pro-resolving mediators (SPMs), probiotics, prebiotics, and certain diets. We believe that successful resolution of chronic inflammation and correction of dysbiosis, in combination with current anti-RA and anti-CC therapies, is a promising therapeutic approach for RA and CC. This approach could also reduce the risk of CC development in HPV-infected RA patients.


Subject(s)
Arthritis, Rheumatoid , Dysbiosis , Papillomavirus Infections , Uterine Cervical Neoplasms , Humans , Uterine Cervical Neoplasms/therapy , Arthritis, Rheumatoid/complications , Female , Dysbiosis/complications , Papillomavirus Infections/complications , Probiotics/therapeutic use , Inflammation , Gastrointestinal Microbiome , Prebiotics , Tumor Microenvironment , Risk Factors
4.
Nature ; 632(8023): 174-181, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987594

ABSTRACT

Changes in the gut microbiome have pivotal roles in the pathogenesis of acute graft-versus-host disease (aGVHD) after allogenic haematopoietic cell transplantation (allo-HCT)1-6. However, effective methods for safely resolving gut dysbiosis have not yet been established. An expansion of the pathogen Enterococcus faecalis in the intestine, associated with dysbiosis, has been shown to be a risk factor for aGVHD7-10. Here we analyse the intestinal microbiome of patients with allo-HCT, and find that E. faecalis escapes elimination and proliferates in the intestine by forming biofilms, rather than by acquiring drug-resistance genes. We isolated cytolysin-positive highly pathogenic E. faecalis from faecal samples and identified an anti-E. faecalis enzyme derived from E. faecalis-specific bacteriophages by analysing bacterial whole-genome sequencing data. The antibacterial enzyme had lytic activity against the biofilm of E. faecalis in vitro and in vivo. Furthermore, in aGVHD-induced gnotobiotic mice that were colonized with E. faecalis or with patient faecal samples characterized by the domination of Enterococcus, levels of intestinal cytolysin-positive E. faecalis were decreased and survival was significantly increased in the group that was treated with the E. faecalis-specific enzyme, compared with controls. Thus, administration of a phage-derived antibacterial enzyme that is specific to biofilm-forming pathogenic E. faecalis-which is difficult to eliminate with existing antibiotics-might provide an approach to protect against aGVHD.


Subject(s)
Bacteriophages , Enterococcus faecalis , Gastrointestinal Microbiome , Graft vs Host Disease , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Young Adult , Bacteriophages/enzymology , Bacteriophages/genetics , Biofilms/drug effects , Biofilms/growth & development , Dysbiosis/complications , Dysbiosis/microbiology , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Enterococcus faecalis/growth & development , Enterococcus faecalis/metabolism , Enterococcus faecalis/virology , Feces/microbiology , Germ-Free Life , Graft vs Host Disease/complications , Graft vs Host Disease/microbiology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , In Vitro Techniques , Intestines/drug effects , Intestines/microbiology , Perforin/metabolism , Risk Factors , Transplantation, Homologous/adverse effects , Whole Genome Sequencing , Drug Resistance, Bacterial/drug effects , Anti-Bacterial Agents/pharmacology
5.
Crit Care ; 28(1): 220, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965622

ABSTRACT

The gut serves as a vital immunological organ orchestrating immune responses and influencing distant mucosal sites, notably the respiratory mucosa. It is increasingly recognized as a central driver of critical illnesses, with intestinal hyperpermeability facilitating bacterial translocation, systemic inflammation, and organ damage. The "gut-lung" axis emerges as a pivotal pathway, where gut-derived injurious factors trigger acute lung injury (ALI) through the systemic circulation. Direct and indirect effects of gut microbiota significantly impact immune responses. Dysbiosis, particularly intestinal dysbiosis, termed as an imbalance of microbial species and a reduction in microbial diversity within certain bodily microbiomes, influences adaptive immune responses, including differentiating T regulatory cells (Tregs) and T helper 17 (Th17) cells, which are critical in various lung inflammatory conditions. Additionally, gut and bone marrow immune cells impact pulmonary immune activity, underscoring the complex gut-lung interplay. Moreover, lung microbiota alterations are implicated in diverse gut pathologies, affecting local and systemic immune landscapes. Notably, lung dysbiosis can reciprocally influence gut microbiota composition, indicating bidirectional gut-lung communication. In this review, we investigate the pathophysiology of ALI/acute respiratory distress syndrome (ARDS), elucidating the role of immune cells in the gut-lung axis based on recent experimental and clinical research. This exploration aims to enhance understanding of ALI/ARDS pathogenesis and to underscore the significance of gut-lung interactions in respiratory diseases.


Subject(s)
Gastrointestinal Microbiome , Respiratory Distress Syndrome , Humans , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Microbiome/immunology , Lung/immunology , Lung/physiopathology , Lung/microbiology , Dysbiosis/physiopathology , Dysbiosis/immunology , Dysbiosis/complications , Animals
6.
BMC Nephrol ; 25(1): 203, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907188

ABSTRACT

IgA nephropathy, presently recognized as the foremost primary glomerular disorder, emerges as a principal contributor to renal failure globally, with its pathogenesis yet to be fully elucidated. Extensive research has highlighted the critical role of gut microbiome in the onset and progression of IgA nephropathy, underscoring its importance in accurately delineating the disease's etiology. For example, gut microbiome dysbacteriosis can lead to the production of nephritogenic IgA1 antibodies, which form immune complexes that deposit in the kidneys, causing inflammation and damage. The gut microbiome, a source of numerous bioactive compounds, interacts with the host and plays a regulatory role in gut-immune axis modulation, earning it the moniker of the "second brain." Recent investigations have particularly emphasized a significant correlation between IgA nephropathy and gut microbiome dysbacteriosis. This article offers a detailed overview of the pathogenic mechanisms of IgA nephropathy, specifically focusing on elucidating how alterations in the gut microbiome are associated with anomalies in the intestinal mucosal system in IgA nephropathy. Additionally, it describes the possible influence of gut microbiome on recurrent IgA nephropathy following kidney transplantation. Furthermore, it compiles potential therapeutic interventions, offering both theoretical and practical foundations for the management of IgA nephropathy. Lastly, the challenges currently faced in the therapeutic approaches to IgA nephropathy are discussed.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Glomerulonephritis, IGA , Immunity, Mucosal , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/microbiology , Humans , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Dysbiosis/complications , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Kidney Transplantation
7.
Life Sci ; 351: 122793, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38848938

ABSTRACT

The enteric nervous system (ENS) regulates numerous functional and immunological attributes of the gastrointestinal tract. Alterations in ENS cell function have been linked to intestinal outcomes in various metabolic, intestinal, and neurological disorders. Chronic kidney disease (CKD) is associated with a challenging intestinal environment due to gut dysbiosis, which further affects patient quality of life. Although the gut-related repercussions of CKD have been thoroughly investigated, the involvement of the ENS in this puzzle remains unclear. ENS cell dysfunction, such as glial reactivity and alterations in cholinergic signaling in the small intestine and colon, in CKD are associated with a wide range of intestinal pathways and responses in affected patients. This review discusses how the ENS is affected in CKD and how it is involved in gut-related outcomes, including intestinal permeability, inflammation, oxidative stress, and dysmotility.


Subject(s)
Enteric Nervous System , Renal Insufficiency, Chronic , Humans , Enteric Nervous System/physiopathology , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/metabolism , Animals , Kidney/physiopathology , Gastrointestinal Microbiome , Oxidative Stress , Dysbiosis/complications , Gastrointestinal Tract/physiopathology , Gastrointestinal Tract/metabolism , Inflammation
8.
Curr Opin Gastroenterol ; 40(5): 413-421, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38900442

ABSTRACT

PURPOSE OF REVIEW: This review evaluates the current knowledge of gut microbiome alterations in acute pancreatitis, including those that can increase acute pancreatitis risk or worsen disease severity, and the mechanisms of gut microbiome driven injury in acute pancreatitis. RECENT FINDINGS: Recent observational studies in humans showed the association of gut microbiome changes (decreased gut microbiome diversity, alterations in relative abundances of certain species, and association of unique species with functional pathways) with acute pancreatitis risk and severity. Furthermore, in-vivo studies highlighted the role of gut microbiome in the development and severity of acute pancreatitis using FMT models. The gut barrier integrity, immune cell homeostasis, and microbial metabolites appear to play key roles in acute pancreatitis risk and severity. SUMMARY: Large human cohort studies that assess gut microbiome profile, its metabolites and impact on acute pancreatitis risk and severity will be crucial for development of innovative prediction, prevention and treatment strategies.


Subject(s)
Gastrointestinal Microbiome , Pancreatitis , Humans , Pancreatitis/microbiology , Gastrointestinal Microbiome/physiology , Severity of Illness Index , Dysbiosis/microbiology , Dysbiosis/complications , Dysbiosis/immunology , Acute Disease , Fecal Microbiota Transplantation
9.
Nature ; 629(8012): 652-659, 2024 May.
Article in English | MEDLINE | ID: mdl-38693261

ABSTRACT

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Subject(s)
Disease Susceptibility , Dysbiosis , Fathers , Gastrointestinal Microbiome , Placental Insufficiency , Prenatal Injuries , Spermatozoa , Animals , Female , Male , Mice , Pregnancy , Dysbiosis/complications , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Leptin/metabolism , Mice, Inbred C57BL , Placenta/metabolism , Placenta/physiopathology , Placental Insufficiency/etiology , Placental Insufficiency/metabolism , Placental Insufficiency/physiopathology , Pregnancy Outcome , Prenatal Injuries/etiology , Prenatal Injuries/metabolism , Prenatal Injuries/physiopathology , Signal Transduction , Spermatozoa/metabolism , Testis/metabolism , Testis/physiopathology , Disease Susceptibility/etiology
10.
Hypertension ; 81(7): 1467-1476, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38716665

ABSTRACT

Epidemiological studies have revealed that hypertensive heart disease is a major risk factor for heart failure, and its heart failure burden is growing rapidly. The need to act in the face of this threat requires first an understanding of the multifactorial origin of hypertensive heart disease and second an exploration of new mechanistic pathways involved in myocardial alterations critically involved in cardiac dysfunction and failure (eg, myocardial interstitial fibrosis). Increasing evidence shows that alterations of gut microbiota composition and function (ie, dysbiosis) leading to changes in microbiota-derived metabolites and impairment of the gut barrier and immune functions may be involved in blood pressure elevation and hypertensive organ damage. In this review, we highlight recent advances in the potential contribution of gut microbiota alterations to myocardial interstitial fibrosis in hypertensive heart disease through blood pressure-dependent and blood pressure-independent mechanisms. Achievements in this field should open a new path for more comprehensive treatment of myocardial interstitial fibrosis in hypertensive heart disease and, thus, for the prevention of heart failure.


Subject(s)
Fibrosis , Gastrointestinal Microbiome , Heart Failure , Hypertension , Myocardium , Humans , Gastrointestinal Microbiome/physiology , Heart Failure/microbiology , Heart Failure/physiopathology , Hypertension/complications , Myocardium/pathology , Myocardium/metabolism , Dysbiosis/microbiology , Dysbiosis/complications , Animals
11.
Indian J Gastroenterol ; 43(4): 729-739, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38767806

ABSTRACT

BACKGROUND: Poor oral health and oral dysbiosis were found to be associated with cancers, especially of the gastrointestinal (GI) system. But the cause-and-effect relationship and the effect of the risk are not yet known due to scarcity of literature. Understanding such risk relationship can contribute to an integrated multi-disciplinary approach for GI cancer prevention. AIM: The aim of the present systematic review and meta-analysis is to assess the role of oral dysbiosis on increasing the risk of digestive system cancers. OBJECTIVE: To evaluate the effect of poor oral health on increasing the risk of gastrointestinal cancers. METHODS: We conducted a systematic search following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines in databases PubMed, Elsevier, Wiley's online library and Web of Science from inception to February 2023 to include recent cohort studies that assessed the association between poor oral health and the risk of cancer. We assessed bias using the New Castle Ottawa scale. We used inferential statistics to describe the effect of oral dysbiosis on gastrointestinal cancers. We performed a sub-group analysis to assess the effect of oral conditions on individual cancers. RESULTS: We included 10 longitudinal studies in the meta-analysis. The overall effect size of poor oral health and GI cancer risk was hazard's ratio (HR) =1.30 (95% CI: [1.14, 1.46]) (p<0.001) (I2 = 68.78). Sub-group analysis indicated that poor oral health increases the risk of esophageal cancer HR=1.61 (95% CI: [1.37, 1.85]), stomach cancer HR=1.33 (95% CI: [1.08, 1.58]), pancreatic cancer HR=1.90 (95% CI; [1.29, 2.50]) and colorectal and hepatocellular carcinoma HR=1.16 (95% CI: [1.08, 1.23]). CONCLUSION: The meta-analysis indicated that poor oral health was significantly associated with increasing the risk of GI cancers.


Subject(s)
Dysbiosis , Gastrointestinal Neoplasms , Oral Health , Humans , Dysbiosis/complications , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/epidemiology , Longitudinal Studies , Risk Factors , Mouth Diseases/epidemiology , Mouth Diseases/etiology , Risk
12.
Medicina (Kaunas) ; 60(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38674238

ABSTRACT

The human microbiome has a crucial role in the homeostasis and health of the host. These microorganisms along with their genes are involved in various processes, among these are neurological signaling, the maturation of the immune system, and the inhibition of opportunistic pathogens. In this sense, it has been shown that a healthy ocular microbiota acts as a barrier against the entry of pathogens, contributing to the prevention of infections. In recent years, a relationship has been suggested between microbiota dysbiosis and the development of neurodegenerative diseases. In patients with glaucoma, it has been observed that the microbiota of the ocular surface, intraocular cavity, oral cavity, stomach, and gut differ from those observed in healthy patients, which may suggest a role in pathology development, although the evidence remains limited. The mechanisms involved in the relationship of the human microbiome and this neurodegenerative disease remain largely unknown. For this reason, the present review aims to show a broad overview of the influence of the structure and composition of the human oral and gut microbiota and relate its dysbiosis to neurodegenerative diseases, especially glaucoma.


Subject(s)
Dysbiosis , Glaucoma , Microbiota , Humans , Glaucoma/microbiology , Microbiota/physiology , Dysbiosis/complications , Dysbiosis/immunology , Mouth/microbiology , Gastrointestinal Microbiome/physiology , Eye/microbiology , Neurodegenerative Diseases/microbiology
13.
Front Immunol ; 15: 1352744, 2024.
Article in English | MEDLINE | ID: mdl-38605969

ABSTRACT

Like other infections, a SARS-CoV-2 infection can also trigger Post-Acute Infection Syndromes (PAIS), which often progress into myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). ME/CFS, characterized by post-exercise malaise (PEM), is a severe multisystemic disease for which specific diagnostic markers or therapeutic concepts have not been established. Despite numerous indications of post-infectious neurological, immunological, endocrinal, and metabolic deviations, the exact causes and pathophysiology remain unclear. To date, there is a paucity of data, that changes in the composition and function of the gastrointestinal microbiota have emerged as a potential influencing variable associated with immunological and inflammatory pathways, shifts in ME/CFS. It is postulated that this dysbiosis may lead to intestinal barrier dysfunction, translocation of microbial components with increased oxidative stress, and the development or progression of ME/CFS. In this review, we detailed discuss the findings regarding alterations in the gastrointestinal microbiota and its microbial mediators in ME/CFS. When viewed critically, there is currently no evidence indicating causality between changes in the microbiota and the development of ME/CFS. Most studies describe associations within poorly defined patient populations, often combining various clinical presentations, such as irritable bowel syndrome and fatigue associated with ME/CFS. Nevertheless, drawing on analogies with other gastrointestinal diseases, there is potential to develop strategies aimed at modulating the gut microbiota and/or its metabolites as potential treatments for ME/CFS and other PAIS. These strategies should be further investigated in clinical trials.


Subject(s)
Fatigue Syndrome, Chronic , Gastrointestinal Diseases , Gastrointestinal Microbiome , Humans , Fatigue Syndrome, Chronic/etiology , Gastrointestinal Diseases/complications , Oxidative Stress , Dysbiosis/complications
14.
Lipids Health Dis ; 23(1): 117, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649999

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) has garnered considerable attention globally. Changing lifestyles, over-nutrition, and physical inactivity have promoted its development. MASLD is typically accompanied by obesity and is strongly linked to metabolic syndromes. Given that MASLD prevalence is on the rise, there is an urgent need to elucidate its pathogenesis. Hepatic lipid accumulation generally triggers lipotoxicity and induces MASLD or progress to metabolic dysfunction-associated steatohepatitis (MASH) by mediating endoplasmic reticulum stress, oxidative stress, organelle dysfunction, and ferroptosis. Recently, significant attention has been directed towards exploring the role of gut microbial dysbiosis in the development of MASLD, offering a novel therapeutic target for MASLD. Considering that there are no recognized pharmacological therapies due to the diversity of mechanisms involved in MASLD and the difficulty associated with undertaking clinical trials, potential targets in MASLD remain elusive. Thus, this article aimed to summarize and evaluate the prominent roles of lipotoxicity, ferroptosis, and gut microbes in the development of MASLD and the mechanisms underlying their effects. Furthermore, existing advances and challenges in the treatment of MASLD were outlined.


Subject(s)
Endoplasmic Reticulum Stress , Ferroptosis , Gastrointestinal Microbiome , Humans , Oxidative Stress , Dysbiosis/complications , Dysbiosis/microbiology , Animals , Fatty Liver/metabolism , Lipid Metabolism , Obesity/metabolism , Obesity/complications , Obesity/pathology , Liver/metabolism , Liver/pathology , Metabolic Syndrome/metabolism
15.
Genome Med ; 16(1): 59, 2024 04 20.
Article in English | MEDLINE | ID: mdl-38643166

ABSTRACT

BACKGROUND: Gut dysbiosis has been linked with both HIV infection and diabetes, but its interplay with metabolic and inflammatory responses in diabetes, particularly in the context of HIV infection, remains unclear. METHODS: We first conducted a cross-sectional association analysis to characterize the gut microbial, circulating metabolite, and immune/inflammatory protein features associated with diabetes in up to 493 women (~ 146 with prevalent diabetes with 69.9% HIV +) of the Women's Interagency HIV Study. Prospective analyses were then conducted to determine associations of identified metabolites with incident diabetes over 12 years of follow-up in 694 participants (391 women from WIHS and 303 men from the Multicenter AIDS Cohort Study; 166 incident cases were recorded) with and without HIV infection. Mediation analyses were conducted to explore whether gut bacteria-diabetes associations are explained by altered metabolites and proteins. RESULTS: Seven gut bacterial genera were identified to be associated with diabetes (FDR-q < 0.1), with positive associations for Shigella, Escherichia, Megasphaera, and Lactobacillus, and inverse associations for Adlercreutzia, Ruminococcus, and Intestinibacter. Importantly, the associations of most species, especially Adlercreutzia and Ruminococcus, were largely independent of antidiabetic medications use. Meanwhile, 18 proteins and 76 metabolites, including 3 microbially derived metabolites (trimethylamine N-oxide, phenylacetylglutamine (PAGln), imidazolepropionic acid (IMP)), 50 lipids (e.g., diradylglycerols (DGs) and triradylglycerols (TGs)) and 23 non-lipid metabolites, were associated with diabetes (FDR-q < 0.1), with the majority showing positive associations and more than half of them (59/76) associated with incident diabetes. In mediation analyses, several proteins, especially interleukin-18 receptor 1 and osteoprotegerin, IMP and PAGln partially mediate the observed bacterial genera-diabetes associations, particularly for those of Adlercreutzia and Escherichia. Many diabetes-associated metabolites and proteins were altered in HIV, but no effect modification on their associations with diabetes was observed by HIV. CONCLUSION: Among individuals with and without HIV, multiple gut bacterial genera, blood metabolites, and proinflammatory proteins were associated with diabetes. The observed mediated effects by metabolites and proteins in genera-diabetes associations highlighted the potential involvement of inflammatory and metabolic perturbations in the link between gut dysbiosis and diabetes in the context of HIV infection.


Subject(s)
Diabetes Mellitus , HIV Infections , Male , Humans , Female , HIV Infections/drug therapy , Prospective Studies , Cohort Studies , Dysbiosis/complications , Cross-Sectional Studies , Bacteria
16.
Curr Opin Obstet Gynecol ; 36(3): 155-164, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38597377

ABSTRACT

PURPOSE OF REVIEW: To examine impact of vaginal dysbiosis (VD), including bacterial vaginosis (BV) and aerobic vaginitis (AV) on reproductive outcomes of in vitro fertilization (IVF) patients. RECENT FINDINGS: BV-bacteria (e.g. Gardnerella ) and AV-bacteria (e.g. Streptococci and Enterococci ) have been identified in the endometrium. However, there is inconclusive evidence whether IVF patients with VD have lower success rates. SUMMARY: The present systematic review and meta-analysis of PubMed/Medline, until December 2023 included 25 studies, involving 6835 IVF patients. Overall VD was defined as an approximation of community state type IV, including BV and AV-type dysbiosis based on either molecular or microscopy methods. Outcomes were live birth rate (LBR), early pregnancy loss (EPL), clinical pregnancy rate (CPR), and biochemical pregnancy rate (BPR).Vaginal dysbiosis prevalence was 19% [1271/6835, 95% confidence interval (CI) 18-20%]. Six studies examined AV-type dysbiosis with a prevalence of 4% (26/628, 95% CI 3-6%). Vaginal dysbiosis correlates with a higher EPL [relative risk (RR) = 1.49, 95% CI 1.15-1.94] and lower CPR (RR = 0.82, 95% CI 0.70-0.95). No statistically significant impact of VD, BV, or AV was found on LBR and BPR.Thus, the association between VD and reproductive outcome remains puzzling as it is difficult to explain how VD impacts CPR and EPL but not LBR and BPR.


Subject(s)
Dysbiosis , Fertilization in Vitro , Pregnancy Rate , Vagina , Vaginosis, Bacterial , Humans , Female , Dysbiosis/complications , Pregnancy , Vaginosis, Bacterial/complications , Vaginosis, Bacterial/microbiology , Vagina/microbiology , Abortion, Spontaneous/microbiology , Pregnancy Outcome , Vaginitis/microbiology , Live Birth
18.
Neurochem Int ; 175: 105724, 2024 May.
Article in English | MEDLINE | ID: mdl-38508416

ABSTRACT

Both Helicobacter pylori (H. pylori) infection and metabolic syndrome (MetS) are highly prevalent worldwide. The emergence of relevant research suggesting a pathogenic linkage between H. pylori infection and MetS-related cardio-cerebrovascular diseases and neurodegenerative disorders, particularly through mechanisms involving brain pericyte deficiency, hyperhomocysteinemia, hyperfibrinogenemia, elevated lipoprotein-a, galectin-3 overexpression, atrial fibrillation, and gut dysbiosis, has raised stimulating questions regarding their pathophysiology and its translational implications for clinicians. An additional stimulating aspect refers to H. pylori and MetS-related activation of innate immune cells, mast cells (MC), which is an important, often early, event in systemic inflammatory pathologies and related brain disorders. Synoptically, MC degranulation may play a role in the pathogenesis of H. pylori and MetS-related obesity, adipokine effects, dyslipidemia, diabetes mellitus, insulin resistance, arterial hypertension, vascular dysfunction and arterial stiffness, an early indicator of atherosclerosis associated with cardio-cerebrovascular and neurodegenerative disorders. Meningeal MC can be activated by triggers including stress and toxins resulting in vascular changes and neurodegeneration. Likewise, H.pylori and MetS-related MC activation is linked with: (a) vasculitis and thromboembolic events that increase the risk of cardio-cerebrovascular and neurodegenerative disorders, and (b) gut dysbiosis-associated neurodegeneration, whereas modulation of gut microbiota and MC activation may promote neuroprotection. This narrative review investigates the intricate relationship between H. pylori infection, MetS, MC activation, and their collective impact on pathophysiological processes linked to neurodegeneration. Through a comprehensive search of current literature, we elucidate the mechanisms through which H. pylori and MetS contribute to MC activation, subsequently triggering cascades of inflammatory responses. This highlights the role of MC as key mediators in the pathogenesis of cardio-cerebrovascular and neurodegenerative disorders, emphasizing their involvement in neuroinflammation, vascular dysfunction and, ultimately, neuronal damage. Although further research is warranted, we provide a novel perspective on the pathophysiology and management of brain disorders by exploring potential therapeutic strategies targeting H. pylori eradication, MetS management, and modulation of MC to mitigate neurodegeneration risk while promoting neuroprotection.


Subject(s)
Brain Diseases , Helicobacter Infections , Helicobacter pylori , Metabolic Syndrome , Neurodegenerative Diseases , Humans , Metabolic Syndrome/complications , Metabolic Syndrome/metabolism , Mast Cells/metabolism , Dysbiosis/complications , Helicobacter Infections/drug therapy , Neurodegenerative Diseases/metabolism
19.
Front Immunol ; 15: 1331486, 2024.
Article in English | MEDLINE | ID: mdl-38510244

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disorder characterized by swollen joints, discomfort, stiffness, osteoporosis, and reduced functionality. Genetics, smoking, dust inhalation, high BMI, and hormonal and gut microbiota dysbiosis are all likely causes of the onset or development of RA, but the underlying mechanism remains unknown. Compared to healthy controls, patients with RA have a significantly different composition of gut microbiota. It is well known that the human gut microbiota plays a key role in the initiation, maintenance, and operation of the host immune system. Gut microbiota dysbiosis has local or systematic adverse effects on the host immune system, resulting in host susceptibility to various diseases, including RA. Studies on the intestinal microbiota modulation and immunomodulatory properties of probiotics have been reported, in order to identify their potential possibility in prevention and disease activity control of RA. This review summarized current studies on the role and potential mechanisms of gut microbiota in the development and progression of RA, as well as the preventative and therapeutic effects and potential mechanisms of probiotics on RA. Additionally, we proposed the challenges and difficulties in the application of probiotics in RA, providing the direction for the research and application of probiotics in the prevention of RA.


Subject(s)
Arthritis, Rheumatoid , Gastrointestinal Microbiome , Probiotics , Humans , Dysbiosis/complications , Arthritis, Rheumatoid/drug therapy , Immune System , Probiotics/therapeutic use
20.
J Gastrointestin Liver Dis ; 33(1): 30-36, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38554421

ABSTRACT

BACKGROUND AND AIMS: The mineral compound Luvos Healing Earth (LHE) is a commercially available remedy empirically used for a variety of gastrointestinal disorders. The aim of this study was to investigate the possible effect of prolonged LHE therapy on gut microbiota in healthy individuals and in patients with diarrhea-predominant irritable bowel syndrome (IBS-D). METHODS: In this prospective exploratory study, a total of 20 participants, including 12 healthy controls and 8 patients with IBS-D, received treatment with LHE (Magenfein Granulat, 1 sachet bid) for 6 weeks. Fecal samples were collected for microbiota analysis in the morning fasting state at regular intervals at 6 different timepoints: 2 weeks before starting therapy (Screen), and every 2 weeks during LHE therapy (V0-V3). Additionally, a follow-up visit was scheduled 4 weeks after the end of treatment (V4). Microbiota analysis was performed using the GA-map® Dysbiosis Test Lx v2. Dysbiosis Index, bacterial diversity, as well as the balance or imbalance of functionally important bacteria were assessed. RESULTS: The microbiota analysis revealed an overlap in gut microbiota profiles between healthy controls and patients with IBS-D. Bacterial communities were consistently stable during the entire treatment period, and no significant variations in composition were observed 4 weeks after the end of the therapeutic intervention. There was a remarkable stability of microbiota profiles over time within each individual and a high inter-individual variation. The majority of fecal samples exhibited profiles, reflecting an eubiotic state, with no significant changes in dysbiosis index, functional bacteria profiles, or bacterial diversity. CONCLUSION: Our findings indicate intraindividual resilience of microbiota consortia during the entire study period. Prolonged intake of LHE does not cause significant alterations in fecal microbiota profiles in healthy controls and patients with IBS-D. Luvos Healing Earth does not affect the stability of gut microbial diversity and bacterial functions.


Subject(s)
Gastrointestinal Microbiome , Irritable Bowel Syndrome , Humans , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/complications , Diarrhea/diagnosis , Diarrhea/etiology , Dysbiosis/complications , Dysbiosis/microbiology , Prospective Studies , Feces/microbiology , Bacteria
SELECTION OF CITATIONS
SEARCH DETAIL