Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1361440, 2024.
Article in English | MEDLINE | ID: mdl-39027478

ABSTRACT

Background: To clarify the causal relationship between gut microbiota and diabetic nephropathy (DN), we employed Mendelian randomization (MR). Despite a strong correlation observed, establishing causality is still unclear. By utilizing MR, we aimed to investigate this relationship further and shed light on the potential causal effect of gut microbiota on DN. Methods: Genetic instrumental variables for gut microbiota were obtained from a GWAS with 18340 participants. DN summary statistics (1032 cases, 451248 controls) were sourced from a separate GWAS. The primary analysis used the inverse-variance weighted (IVW) method. Reverse MR analysis was conducted to explore reverse causation. Rigorous sensitivity analyses were performed to ensure the resilience and reliability of the study's findings. Results: We found two bacterial traits associated with an increased risk of DN: genus LachnospiraceaeUCG008 (OR: 1.4210; 95% CI: 1.0450, 1.9322; p = 0.0250) and genus Terrisporobacter (OR: 1.9716; 95% CI: 1.2040, 3.2285; p = 0.0070). Additionally, phylum Proteobacteria (OR: 0.4394; 95% CI: 0.2721, 0.7096; p = 0.0008) and genus Dialister (OR: 0.4841; 95% CI: 0.3171, 0.7390; p = 0.0008) were protective against DN. Sensitivity analyses consistently supported these results. In the reverse MR analysis, no statistically significant associations were observed between DN and these four bacterial traits. Conclusions: Our analyses confirmed a potential causal relationship between certain gut microbiota taxa and the risk of DN. However, additional studies are required to elucidate the underlying mechanisms through which gut microbiota influences the development of DN.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Humans , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/genetics
2.
Diabetes Res Clin Pract ; 213: 111726, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844054

ABSTRACT

Diabetic nephropathy is a common complication of diabetes and a considerable contributor to end-stage renal disease. Evidence indicates that glucose dysregulation and lipid metabolism comprise a pivotal pathogenic mechanism in diabetic nephropathy. However, current treatment outcomes are limited, as they only provide symptomatic relief without preventing disease progression. The gut microbiota is a group of microorganisms that inhabit the human intestinal tract and play a crucial role in maintaining host energy balance, metabolism, and immune activity. Patients with diabetic nephropathy exhibit altered gut microbiota, suggesting its potential involvement in the onset and progression of the disease. However, how a perturbed microbiota induces and promotes diabetic nephropathy remains unelucidated. This article summarizes the evidence of the impact of gut microbiota on the progression of diabetic nephropathy, with a particular focus on the molecular mechanisms involved, aiming to provide new insights into the treatment of diabetic nephropathy.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/metabolism , Humans , Gastrointestinal Microbiome/physiology , Animals , Disease Progression
3.
Ren Fail ; 46(2): 2357746, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38832498

ABSTRACT

Numerous studies have revealed a correlation between the risk of developing diabetic nephropathy (DN) and the gut microbiota (GM) composition. However, it remains uncertain whether the GM composition causes DN. We aimed to explore any potential causal links between the GM composition and the risk of developing DN. A meta-analysis conducted by the MiBioGen consortium of the largest genome-wide association study (GWAS) provided aggregated data on the GM. DN data were obtained from the IEU database. The inverse-variance weighting (IVW) method was employed as the primary analytical approach. The IVW analysis indicated that genus Dialister (OR = 0.51, 95% CI: 0.34-0.77, p = 0.00118) was protective against DN. In addition, class Gammaproteobacteria (OR = 0.47, 95% CI: 0.27-0.83, p = 0.0096), class Lentisphaeria (OR =0.76, 95% CI: 0.68-0.99, p = 0.04), order Victivallales (OR = 0.76, 95% CI: 0.58-0.99, p = 0.04), and phylum Proteobacteria (OR = 0.53, 95% CI: 0.33-0.85, p = 0.00872) were negatively associated with the risk of developing DN. Genus LachnospiraceaeUCG008 (OR =1.45, 95% CI: 1.08-1.95, p = 0.01), order Bacteroidales (OR = 1.59, 95% CI: 1.02-2.49, p = 0.04), and genus Terrisporobacter (OR = 1.98, 95% CI: 1.14-3.45, p = 0.015) were positively associated with the risk of developing DN. In this study, we established a causal relationship between the genus Dialister and the risk of developing DN. Further trials are required to confirm the protective effects of probiotics on DN and to elucidate the precise protective mechanisms involving genus Dialister and DN.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Humans , Diabetic Nephropathies/microbiology , Gastrointestinal Microbiome/genetics
4.
mBio ; 15(7): e0073524, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38819146

ABSTRACT

Current microbiome signatures for chronic diseases such as diabetic kidney disease (DKD) are mainly based on low-resolution taxa such as genus or phyla and are often inconsistent among studies. In microbial ecosystems, bacterial functions are strain specific, and taxonomically different bacteria tend to form co-abundance functional groups called guilds. Here, we identified guild-level signatures for DKD by performing in-depth metagenomic sequencing and conducting genome-centric and guild-based analysis on fecal samples from 116 DKD patients and 91 healthy subjects. Redundancy analysis on 1,543 high-quality metagenome-assembled genomes (HQMAGs) identified 54 HQMAGs that were differentially distributed among the young healthy control group, elderly healthy control group, early-stage DKD patients (EDG), and late-stage DKD patients (LDG). Co-abundance network analysis classified the 54 HQMAGs into two guilds. Compared to guild 2, guild 1 contained more short-chain fatty acid biosynthesis genes and fewer genes encoding uremic toxin indole biosynthesis, antibiotic resistance, and virulence factors. Guild indices, derived from the total abundance of guild members and their diversity, delineated DKD patients from healthy subjects and between different severities of DKD. Age-adjusted partial Spearman correlation analysis showed that the guild indices were correlated with DKD disease progression and with risk indicators of poor prognosis. We further validated that the random forest classification model established with the 54 HQMAGs was also applicable for classifying patients with end-stage renal disease and healthy subjects in an independent data set. Therefore, this genome-level, guild-based microbial analysis strategy may identify DKD patients with different severity at an earlier stage to guide clinical interventions. IMPORTANCE: Traditionally, microbiome research has been constrained by the reliance on taxonomic classifications that may not reflect the functional dynamics or the ecological interactions within microbial communities. By transcending these limitations with a genome-centric and guild-based analysis, our study sheds light on the intricate and specific interactions between microbial strains and diabetic kidney disease (DKD). We have unveiled two distinct microbial guilds with opposite influences on host health, which may redefine our understanding of microbial contributions to disease progression. The implications of our findings extend beyond mere association, providing potential pathways for intervention and opening new avenues for patient stratification in clinical settings. This work paves the way for a paradigm shift in microbiome research in DKD and potentially other chronic kidney diseases, from a focus on taxonomy to a more nuanced view of microbial ecology and function that is more closely aligned with clinical outcomes.


Subject(s)
Bacteria , Diabetic Nephropathies , Feces , Gastrointestinal Microbiome , Metagenome , Metagenomics , Humans , Gastrointestinal Microbiome/genetics , Diabetic Nephropathies/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Male , Female , Feces/microbiology , Middle Aged , Adult , Aged
5.
Front Cell Infect Microbiol ; 14: 1359432, 2024.
Article in English | MEDLINE | ID: mdl-38779567

ABSTRACT

Diabetic nephropathy (DN) is one of the main complications of diabetes and a major cause of end-stage renal disease, which has a severe impact on the quality of life of patients. Strict control of blood sugar and blood pressure, including the use of renin-angiotensin-aldosterone system inhibitors, can delay the progression of diabetic nephropathy but cannot prevent it from eventually developing into end-stage renal disease. In recent years, many studies have shown a close relationship between gut microbiota imbalance and the occurrence and development of DN. This review discusses the latest research findings on the correlation between gut microbiota and microbial metabolites in DN, including the manifestations of the gut microbiota and microbial metabolites in DN patients, the application of the gut microbiota and microbial metabolites in the diagnosis of DN, their role in disease progression, and so on, to elucidate the role of the gut microbiota and microbial metabolites in the occurrence and prevention of DN and provide a theoretical basis and methods for clinical diagnosis and treatment.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/microbiology , Disease Progression , Dysbiosis
6.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 127-133, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678614

ABSTRACT

In this study, we summarized the key findings and potential implications of association studies investigating the relationship between gut microbiota composition and risks for Diabetic nephropathy (DN). We used Mendelian randomization (MR) analysis to explore the relationship between gut microbiota and DN using two different publicly available DN databases. The results were also summarized using five mainstream MR analysis methods. We controlled for various possible biases in the results. The results showed that specific bacterial genera were associated with increased or decreased risk of DN. These associations can be attributed to a variety of factors, including metabolites produced by certain bacteria. Most of our findings are consistent with the existing research findings, but there are still some differences with the existing results. In addition, we also pointed out that some microbiota that may be associated with DN but remain unnoticed can bring new research directions. Our work made use of MR, a reliable technique for examining causal correlations using genetic data investigating potential processes, carrying out longitudinal studies, looking into intervention options, and using a multi-omics approach may be future research avenues. Further, our findings also point to a few unexplored possible study paths for DN in the future. These initiatives may improve our reconciliation of the internal relationships between the gut microbiota and DN and pave the way for more precise prevention and treatment methods. However, it is also critical to recognize any potential restrictions, such as those caused by sample size, population variety, and analytical techniques.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Mendelian Randomization Analysis , Humans , Gastrointestinal Microbiome/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/microbiology , Risk Factors
7.
Microbiol Spectr ; 11(4): e0038223, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37341590

ABSTRACT

Many studies have suggested that gut microbiota dysbiosis may be one of the pathogenesis factors of diabetes mellitus (DM), while it is not clear whether it is involved in the development of diabetic kidney diseases (DKD). The objective of this study was to determine bacterial taxa biomarkers during the progression of DKD by investigating bacterial compositional changes in early and late DKD. 16S rRNA gene sequencing was performed on fecal samples, including the diabetes mellitus (DM), DNa (early DKD), and DNb (late DKD) groups. Taxonomic annotation of microbial composition was performed. Samples were sequenced on the Illumina NovaSeq platform. At the genus level, we found counts of Fusobacterium, Parabacteroides, and Ruminococcus_gnavus were significantly elevated both in the DNa group (P = 0.0001, 0.0007, and 0.0174, respectively) and the DNb group (P < 0.0001, 0.0012, and 0.0003, respectively) compared with those in the DM group. Only the level of Agathobacter was significantly decreased in the DNa group than the DM group and in the DNb group than the DNa group. Counts of Prevotella_9, Roseburia were significantly decreased in the DNa group compared with those in the DM group (P = 0.001 and 0.006, respectively) and in the DNb group compared with those in the DM group (P < 0.0001 and 0.003, respectively). Levels of Agathobacter, Prevotella_9, Lachnospira, and Roseburia were positively correlated with an estimated glomerular filtration rate (eGFR), but negatively correlated with microalbuminuria (MAU), 24 h urinary protein quantity (24hUP), and serum creatinine (Scr). Moreover, the areas under the curve (AUCs) of Agathobacter and Fusobacteria were 83.33% and 80.77%, respectively, for the DM and DNa cohorts, respectively. Notably, the largest AUC for DNa and DNb cohorts was also that of Agathobacter at 83.60%. Gut microbiota dysbiosis was found in the early and late stages of DKD, especially in the early stage. Agathobacter may be the most promising intestinal bacteria biomarker that can help distinguish different stages of DKD. IMPORTANCE It is not clear as to whether gut microbiota dysbiosis is involved in the progression of DKD. This study may be the first to explore gut microbiota compositional changes in diabetes, early-DKD, and late DKD. We identify different gut microbial characteristics during different stages of DKD. Gut microbiota dysbiosis is found in the early and late stages of DKD. Agathobacter may be the most promising intestinal bacteria biomarker that can help distinguish different stages of DKD, although further studies are warranted to illustrate these mechanisms.


Subject(s)
Diabetic Nephropathies , Gastrointestinal Microbiome , Diabetic Nephropathies/microbiology , Humans , Male , Female , Middle Aged , Clostridiales/isolation & purification , Biomarkers , Diabetes Mellitus , Bacteria/classification , Bacteria/isolation & purification , Feces/microbiology , Kidney Failure, Chronic/microbiology
8.
Microbiol Spectr ; 10(4): e0032422, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35863004

ABSTRACT

Diabetic nephropathy (DN) is the primary cause of end-stage renal disease. Accumulating studies have implied a critical role for the gut microbiota in diabetes mellitus (DM) and DN. However, the precise roles and regulatory mechanisms of the gut microbiota in the pathogenesis of DN remain largely unclear. In this study, metagenomics sequencing was performed using fecal samples from healthy controls (CON) and type 2 diabetes mellitus (T2DM) patients with or without DN. Fresh fecal samples from 15 T2DM patients without DN, 15 DN patients, and 15 age-, gender-, and body mass index (BMI)-matched healthy controls were collected. The compositions and potential functions of the gut microbiota were estimated. Although no difference of gut microbiota α and ß diversity was observed between the CON, T2DM, and DN groups, the relative abundances of butyrate-producing bacteria (Clostridium, Eubacterium, and Roseburia intestinalis) and potential probiotics (Lachnospira and Intestinibacter) were significantly reduced in T2DM and DN patients. Besides, Bacteroides stercoris was significantly enriched in fecal samples from patients with DN. Moreover, Clostridium sp. 26_22 was negatively associated with serum creatinine (P < 0.05). DN patients could be accurately distinguished from CON by Clostridium sp. CAG_768 (area under the curve [AUC] = 0.941), Bacteroides propionicifaciens (AUC = 0.905), and Clostridium sp. CAG_715 (AUC = 0.908). DN patients could be accurately distinguished from T2DM patients by Pseudomonadales, Fusobacterium varium, and Prevotella sp. MSX73 (AUC = 0.889). Regarding the potential bacterial functions of the gut microbiota, the citrate cycle, base excision repair, histidine metabolism, lipoic acid metabolism, and bile acid biosynthesis were enriched in DN patients, while selenium metabolism and branched-chain amino acid biosynthesis were decreased in DN patients. IMPORTANCE Gut microbiota imbalance is found in fecal samples from DN patients, in which Roseburia intestinalis is significantly decreased, while Bacteroides stercoris is increased. There is a significant correlation between gut microbiota imbalance and clinical indexes related to lipid metabolism, glucose metabolism, and renal function. The gut microbiota may be predictive factors for the development and progression of DN, although further studies are warranted to illustrate their regulatory mechanisms.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Gastrointestinal Microbiome , Bacteroides , Clostridiales , Diabetes Mellitus, Type 2/microbiology , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/pathology , Humans
9.
J Med Microbiol ; 71(5)2022 May.
Article in English | MEDLINE | ID: mdl-35617337

ABSTRACT

The occurrence and development of diabetic nephropathy (DN) are closely related to gut microbiota. Paecilomyces cicadae is a medicinal and edible fungus. Radix astragali is a therapeutic material for unifying Chinese Qi. They can delay the occurrence and development of kidney disease. In recent years, solid-state fermentation of edible fungi and traditional Chinese medicine has become a hot issue.Hypothesis/Gap Statement. We assumed that solid-state fermentation products of R. astragali and Paecilomyces cicadidae (RPF) could ameliorate diabetic nephropathy and modulate gut microbiota composition. We aimed to study the function and mechanism of the RPF for ameliorating DN in mice. We investigated the effect of the potential roles of RPF in DN mice and interaction between DN and gut microbiota using animal experiments and gut microbiota measurements. We found that RPF dramatically reduced urine protein, serum creatinine and blood urea nitrogen in DN mice. Furthermore, RPF ameliorated the physiological condition of DN mice by regulating the abundance of intestinal microbiota such as Ruminococcaceae_UCG-014, Allobaculum, Unclassified_f__Lachnospiraceae Alloprevotella and Bacteroides. RPF can ameliorate diabetic nephropathy and modulate gut microbiota composition.


Subject(s)
Cordyceps , Diabetes Mellitus , Diabetic Nephropathies , Gastrointestinal Microbiome , Animals , Astragalus propinquus , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/microbiology , Drugs, Chinese Herbal , Mice
10.
J Clin Lab Anal ; 35(12): e24062, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34689373

ABSTRACT

BACKGROUND: Previous studies found the dysbiosis of intestinal microbiota in diabetic kidney disease (DKD), especially the decreased SCFA-producing bacteria. We aimed to investigate the concentration of the stool and serum short-chain fatty acids (SCFAs), gut microbiota-derived metabolites, in individuals with DKD and reveal the correlations between SCFAs and renal function. METHODS: A total of 30 participants with DKD, 30 participants with type 2 diabetes mellitus (DM), and 30 normal controls (NC) in HwaMei Hospital were recruited from 1/1/2018 to 12/31/2019. Participants with DKD were divided into low estimated glomerular filtration rate (eGFR)(eGFR<60ml/min, n=14) and high eGFR (eGFR≥60ml/min, n=16) subgroups. Stool and serum were measured for SCFAs with gas chromatograph-mass spectrometry. RESULTS: The DKD group showed markedly lower levels of fecal acetate, propionate, and butyrate versus NC (p<0.001, p<0.001, p=0.018, respectively) [1027.32(784.21-1357.90)]vs[2064.59(1561.82-2637.44)]µg/g,[929.53(493.65-1344.26)]vs[1684.57(1110.54-2324.69)]µg/g,[851.39(409.57-1611.65)] vs[1440.74(1004.15-2594.73)]µg/g, respectively, and the lowest fecal total SCFAs concentration among the groups. DKD group also had a lower serum caproate concentration than that with diabetes (p=0.020)[0.57(0.47-0.61)]vs[0.65(0.53-0.79)]µmol/L. In the univariate regression analysis, fecal and serum acetate correlated with eGFR (OR=1.013, p=0.072; OR=1.017, p=0.032). The correlation between serum total SCFAs and eGFR showed statistical significance (OR=1.019, p=0.024) unadjusted and a borderline significance (OR=1.024, p=0.063) when adjusted for Hb and LDL. The decrease in serum acetate and total SCFAs were found of borderline significant difference in both subgroups (p=0.055, p=0.050). CONCLUSION: This study provides evidence that in individuals with DKD, serum and fecal SCFAs levels (fecal level in particular) were lowered, and there was a negative correlation between SCFAs and renal function.


Subject(s)
Diabetic Nephropathies/metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/physiology , Adult , Aged , Case-Control Studies , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/microbiology , Diabetic Nephropathies/microbiology , Fatty Acids, Volatile/analysis , Fatty Acids, Volatile/blood , Feces/microbiology , Female , Gas Chromatography-Mass Spectrometry , Humans , Male , Middle Aged
11.
Nephrology (Carlton) ; 26(8): 641-649, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33715272

ABSTRACT

This review discusses the influence of gut microbiota dysbiosis on diabetic kidney disease through metabolite profile changes and immune and inflammatory mechanisms. We also elaborate on the mechanism of dysbiosis in the onset and development of other kidney diseases.


Subject(s)
Diabetic Nephropathies/microbiology , Dysbiosis/complications , Gastrointestinal Microbiome , Humans
12.
Biomed Pharmacother ; 133: 111061, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378964

ABSTRACT

QiDiTangShen granules (QDTS), a traditional Chinese herbal medicine, have been used in clinical practice for treating diabetic kidney disease for several years. In our previous study, we have demonstrated that QDTS displayed good efficacy on reducing proteinuria in mice with diabetic nephropathy (DN). However, the exact mechanism by which QDTS exerts its reno-protection remains largely unknown. To ascertain whether QDTS could target the gut microbiota-bile acid axis, the db/db mice were adopted as a mouse model of DN. After a 12-week of treatment, we found that QDTS significantly reduced urinary albumin excretion (UAE), and attenuated the pathological injuries of kidney in the db/db mice, while the body weight and blood glucose levels of those mice were not affected. In addition, we found that QDTS significantly altered the gut microbiota composition, and decreased serum levels of total bile acid (TBA) and BA profiles such as ß-muricholic acid (ß-MCA), taurocholic acid (TCA), tauro ß-muricholic acid (Tß-MCA) and deoxycholic acid (DCA). These BAs are associated with the activation of farnesoid X receptor (FXR), which is highly expressed in kidney. However, there was no significant difference between QDTS-treated and -untreated db/db mice regarding the renal expression of FXR, indicating that other mechanisms may be involved. Conclusively, our study revealed that QDTS significantly alleviated renal injuries in mice with DN. The gut microbiota-bile acid axis may be an important target for the reno-protection of QDTS in DN, but the specific mechanism merits further study.


Subject(s)
Bile Acids and Salts/blood , Diabetic Nephropathies/drug therapy , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Kidney/drug effects , Animals , Biomarkers/blood , Diabetic Nephropathies/blood , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/pathology , Disease Models, Animal , Dysbiosis , Feces/microbiology , Intestines/microbiology , Intestines/pathology , Kidney/metabolism , Kidney/ultrastructure , Male , Proteinuria/blood , Proteinuria/microbiology , Proteinuria/prevention & control
13.
Ren Fail ; 42(1): 1100-1110, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33121301

ABSTRACT

BACKGROUND: Diabetic kidney disease (DKD) and membranous nephropathy (MN) are the two major causes of end-stage renal disease (ESRD). Increasing evidence has shown that intestinal dysbiosis is associated with many diseases. The aim of this study was to explore the composition of the gut microbiome in DKD and MN patients. METHODS: 16S rRNA gene sequencing was performed on 271 fecal samples (DKD = 129 and MN = 142), and taxonomic annotation of microbial composition and function was completed. RESULTS: We observed distinct microbial communities between the two groups, with MN samples exhibiting more severe dysbiosis than DKD samples. Relative increases in genera producing short-chain fatty acids (SCFAs) in DKD and a higher proportion of potential pathogens in MN were the main contributors to the microbiome alterations in the two groups. Five-fold cross-validation was performed on a random forest model, and four operational taxonomic unit (OTU)-based microbial markers were selected to distinguish DKD from MN. The results showed 92.42% accuracy in the training set and 94.52% accuracy in the testing set, indicating high potential for these microbiome-based markers in separating MN from DKD. Overexpression of several amino acid metabolic pathways, carbohydrate metabolism and lipid metabolism was found in DKD, while interconversion of pentose/glucoronate and membrane transport in relation to ABC transporters and the phosphotransferase system were increased in MN. CONCLUSION: The composition of the gut microbiome appears to differ considerably between patients with DKD and those with MN. Thus, microbiome-based markers could be used as an alternative tool to distinguish DKD and MN.


Subject(s)
Diabetic Nephropathies/microbiology , Feces/microbiology , Gastrointestinal Microbiome/genetics , Glomerulonephritis, Membranous/microbiology , RNA, Ribosomal, 16S/genetics , Diabetic Nephropathies/diagnosis , Diagnosis, Differential , Dysbiosis/microbiology , Glomerulonephritis, Membranous/diagnosis , Humans , ROC Curve
14.
J Mycol Med ; 30(3): 101009, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32620497

ABSTRACT

Chronic renal disease patients under chronic dialysis (CRDD) have a multifactorial immunological deterioration with an increased risk of Candida infections. Incidence of Candida infections is increasing. Choice of suitable antifungal agents is limited due to the resistance of some species to several antifungals. Aim of the present study was to identify the distribution and antifungal susceptibility patterns of oral isolated Candida species from infected and colonized patients, as well as to investigate the risk factors for oral infection in patients on dialysis. Cross-sectional study, approved by the institutional bioethics committees was performed in CRDD patients. Demographic, clinic data, and oral mucosa samples were obtained. Infection diagnosis was established clinically and confirmed with exfoliative cytology, each sample was plated on CHROMagar Candida and incubated at 36°C for 2 days. Yeast species were identified by carbohydrate assimilation ID 32C AUX system and the apiweb database. For the antifungal susceptibility test, the M44 A-3 method (CLSI) using fluconazole (FCZ), miconazole (MCZ), nystatin (NYS), and voriconazole (VCZ). Study included 119 participants, the main cause of CRD was nephropathy due to DM2 (58%), and three-fourths of the patients were under hemodialysis. Candida prevalence was 56.3% of 67 colonized or infected patients, 88 isolates were obtained. Principal identified species were C. albicans (51.1%), C. glabrata (25%), and C. tropicalis (14.8%). C. glabrata showed a reduced response to FCZ in 50% of isolates and C. albicans had a reduced response in 16% of the isolates. Antifungal agent with the least efficacious response or with the lowest susceptibility in the isolates of these patients was MCZ, followed by VCZ and FCZ, whereas NYS induced the best antifungal response.


Subject(s)
Antifungal Agents/pharmacology , Candida/drug effects , Candida/isolation & purification , Candidiasis, Oral/microbiology , Mouth/microbiology , Renal Insufficiency, Chronic/microbiology , Adult , Aged , Aged, 80 and over , Candida/classification , Candidiasis, Oral/complications , Candidiasis, Oral/diagnosis , Candidiasis, Oral/epidemiology , Cross-Sectional Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/therapy , Diabetic Nephropathies/epidemiology , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/therapy , Drug Resistance, Fungal/drug effects , Female , Humans , Male , Mexico/epidemiology , Microbial Sensitivity Tests , Middle Aged , Renal Dialysis/adverse effects , Renal Dialysis/statistics & numerical data , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/therapy , Young Adult
15.
Biomed Pharmacother ; 129: 110325, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32535383

ABSTRACT

Growing evidence shows that diabetic kidney disease (DKD) is linked with intestinal dysbiosis from gut-derived toxins. Tangshen Formula (TSF) is a traditional Chinese herbal medicine that has been used to treat DKD. In this study, streptozotocin injection and uninephrectomy-induced diabetic nephropathy (DN) rat model was established to explore the impact of TSF on gut microbiota composition, gut-derived toxins, and the downstream inflammatory pathway of urotoxins in the kidney. TSF treatment for 12 weeks showed significant attenuation of both renal histologic injuries and urinary excretion of albumin compared with DN rats without treatment. TSF treatment also reconstructed gut dysbiosis and reduced levels of indoxyl sulfate and metabolic endotoxemia/lipopolysaccharide. MCP-1 and TNF-α were decreased by TSF both in the serum and kidney. In addition, we revealed that the inhibitory effect of TSF on renal inflammation was associated with the inhibition of aryl hydrocarbon, a receptor of indoxyl sulfate, and TLR4, thereby inhibiting JNK and NF-κB signaling in the kidney. Spearman correlation analysis found that a cluster of gut bacterial phyla and genera were significantly correlated with renal pathology, renal function, and systemic inflammation. In conclusion, orally administered TSF significantly inhibited diabetic renal injury, and modulated gut microbiota, which decreased levels of lipopolysaccharide and indoxyl sulfate, and attenuated renal inflammation. Our results indicate that TSF may be used as an agent in the prevention of gut dysbiosis and elimination of intestinal toxins in DN individuals.


Subject(s)
Bacteria/drug effects , Diabetic Nephropathies/prevention & control , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Intestines/drug effects , Kidney/metabolism , Animals , Bacteria/classification , Bacteria/metabolism , Cytokines/blood , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/pathology , Disease Models, Animal , Dysbiosis , Indican/blood , Inflammation Mediators/blood , Intestines/microbiology , Kidney/pathology , Lipopolysaccharides/blood , Male , Rats, Wistar
16.
Theranostics ; 10(6): 2803-2816, 2020.
Article in English | MEDLINE | ID: mdl-32194836

ABSTRACT

Background: Our previous study demonstrated that the disruption of cholesterol homeostasis promotes tubulointerstitial injury in diabetic nephropathy (DN). This study aimed to further investigate the effects of gut microbiota dysbiosis on this process and explored its potential mechanism. Methods: Diabetic rats treated with broad-spectrum oral antibiotics or faecal microbiota transplantation (FMT) from the healthy donor group and human kidney 2 (HK-2) cells stimulated with sodium acetate were used to observe the effects of gut microbiota on cholesterol homeostasis. The gut microbiota distribution was measured by 16S rDNA sequencing with faeces. Serum acetate level was examined by gas chromatographic analysis. Protein expression of G protein coupled receptor 43 (GPR43) and molecules involved in cholesterol homeostasis were assessed by immunohistochemical staining, immunofluorescence staining, and Western Blotting. Results: Depletion of gut microbiota significantly attenuated albuminuria and tubulointerstitial injury. Interestingly, serum acetate levels were also markedly decreased in antibiotics-treated diabetic rats and positively correlated with the cholesterol contents in kidneys. An in vitro study demonstrated that acetate significantly increased cholesterol accumulation in HK-2 cells, which was caused by increased expression of proteins mainly modulating cholesterol synthesis and uptake. As expected, FMT effectively decreased serum acetate levels and alleviated tubulointerstitial injury in diabetic rats through overriding the disruption of cholesterol homeostasis. Furthermore, GPR43 siRNA treatment blocked acetate-mediated cholesterol homeostasis dysregulation in HK-2 cells through decreasing the expression of proteins governed cholesterol synthesis and uptake. Conclusion: Our studies for the first time demonstrated that the acetate produced from gut microbiota mediated the dysregulation of cholesterol homeostasis through the activation of GPR43, thereby contributing to the tubulointerstitial injury of DN, suggesting that gut microbiota reprogramming might be a new strategy for DN prevention and therapy.


Subject(s)
Cholesterol/metabolism , Diabetic Nephropathies , Dysbiosis , Gastrointestinal Microbiome , Nephritis, Interstitial , Acetates/blood , Animals , Cell Line , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/microbiology , Dysbiosis/metabolism , Dysbiosis/microbiology , Homeostasis , Humans , Male , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/microbiology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
17.
Curr Clin Pharmacol ; 15(3): 234-242, 2020.
Article in English | MEDLINE | ID: mdl-32124701

ABSTRACT

BACKGROUND: Diabetic Nephropathy is a frequent complication of diabetes mellitus due to functional and structural modifications in multiple kidney compartments. Probiotics have risen lately as a forthcoming therapeutic intervention but they have not been systematically evaluated in diabetic nephropathy so far. The aim of this systematic review was to evaluate randomized controlled trials and experimental studies assessing the effect of probiotic supplements on diabetic nephropathy. METHODS: An extensive literature search was conducted through electronic databases (PubMed, Scopus, Cinahl and Medline) with the Medical Subject Headings and entry terms of "diabetic nephropathy", "diabetic renal disease" and "probiotics". The search yielded 116 results, 9 of which met the inclusion criteria for this systematic review. RESULTS: Most of the microorganisms used in the studies belonged to the Lactobacillus and Bifidobacterium genus. The dosage ranged from 2×107 to 6×1010 CFU/ g. The form of the probiotics varied across the studies (capsules, sachets, soy milk, kefir and honey). The majority of the studies demonstrated the benefits of probiotic supplementation on the reduction of inflammation, oxidative stress and on the amelioration of renal function biomarkers in subjects with diabetic nephropathy. No major gastrointestinal adverse events were observed during the intervention time with probiotics. CONCLUSION: Findings of this systematic review demonstrate the positive impact of probiotics on Diabetic Nephropathy without any major adverse events. Moreover, future larger randomized controlled trials with bigger samples and longer follow-up time are deemed necessary for further valid results on the effectiveness of probiotic supplementation on Diabetic Nephropathy.


Subject(s)
Diabetic Nephropathies/therapy , Probiotics/administration & dosage , Animals , Bifidobacterium , Diabetic Nephropathies/microbiology , Diabetic Nephropathies/physiopathology , Humans , Inflammation/microbiology , Inflammation/therapy , Lactobacillus , Oxidative Stress , Randomized Controlled Trials as Topic
18.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165764, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32169506

ABSTRACT

Recent studies have shown that laboratory murine autoimmunity models under the same environment display different outcomes. We established diabetic nephropathy model mice under the same environment using the classic streptozotocin method. Renal dysfunction was different among the mice. Proteinuria was more significant in the severe proteinuria group (SP) than in the mild proteinuria group (MP). We hypothesized a role for the gut microbiota in the outcome and reproducibility of induced DN models. 16S rDNA gene sequencing technology was used to analyze the differences in the gut microbiota between the two groups. Here, through fecal microbiota transplantation (FMT) and gas chromatography mass spectrometry (GC-MS), we verified the role of the gut microbiota and its short-chain fatty acid (SCFA) generation in DN mouse renal dysfunction. In the SP group, there was a reduced abundance of Firmicutes (P < 0.0001), and the dominant genus Allobaculum [linear discriminant analysis (LDA) >3, P < 0.05] was positively correlated with body weight (Rho = 0.767, P < 0.01) and blood glucose content (Rho = 0.648, P < 0.05), while the dominant genus Anaerosporobacter (LDA > 3, P < 0.05) was positively correlated with 24-hour urinary protein content (Rho = 0.773, P < 0.01). In the MP group, the dominant genus Blautia (LDA > 3, P < 0.05) was negatively correlated with 24-hour urinary protein content (Rho = -0.829, P < 0.05). The results indicated that Allobaculum and Anaerosporobacter may worsen renal function, while Blautia may be a protective factor in DN. These findings suggested that the gut microbiota may contribute to the heterogeneity of the induced response since we observed potential disease-associated microbial taxonomies and correlations with DN.


Subject(s)
Diabetic Nephropathies/microbiology , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/genetics , Kidney/metabolism , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/pathogenicity , Blood Glucose/genetics , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/pathology , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Humans , Kidney/microbiology , Kidney/pathology , Mice , RNA, Ribosomal, 16S/genetics , Severity of Illness Index , Streptozocin/toxicity
19.
Int J Biol Macromol ; 132: 1001-1011, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30946910

ABSTRACT

Dysbiosis of gut microbiota and low grade inflammation has gradually become a highly potential therapeutic agent for diabetic nephropathy (DN). It has been reported that a large number of polysaccharides have positive effects on DN, including Bupleurum polysaccharides. However, the mechanism remained unclear. This study selected two Bupleurum polysaccharides from different origins to investigate the potential relationship between kidney and gut. Diabetic mice model was established by streptozotocin (STZ, 100 mg/kg) and the treatment groups were treated with two Bupleurum polysaccharides (60 mg/kg) for 6 weeks, respectively. The results showed that the administration of Bupleurum polysaccharides ameliorated diabetic nephropathy induced by STZ. Blood glucose, blood creatinine and urine albumin were decreased after the oral administration of Bupleurum polysaccharides. And the dysbiosis of gut microbiota was modulated with higher diversity and gut protective microbiota. The gut barrier was also improved and the expression of inflammatory response both in kidney and colon was reduced. These results provided the evidence that modulating the gut microbiota and inflammation was involved in the effect of Bupleurum polysaccharides against diabetic nephropathy in mice and laid the foundation for the deeper, specific mechanism research on the interaction between kidney and gut.


Subject(s)
Bupleurum/chemistry , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/microbiology , Gastrointestinal Microbiome/drug effects , Polysaccharides/pharmacology , Animals , Colon/drug effects , Colon/microbiology , Colon/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Glucose/metabolism , Inflammation/drug therapy , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Polysaccharides/therapeutic use
20.
Acta Diabetol ; 56(5): 581-592, 2019 May.
Article in English | MEDLINE | ID: mdl-30888537

ABSTRACT

AIMS: Type 2 diabetes mellitus (T2DM) has a rising prevalence and gut microbiota involvement is increasingly recognized. Diabetic nephropathy (DN) is a major complication of T2DM. The aim of the study was to understand the gut-kidney axis by an analysis of gut microbiota composition among biopsy-proven DN, T2DM without kidney disease, and healthy control. METHODS: Fecal samples were collected from 14 DNs, 14 age/gender-matched T2DMs without renal diseases (DM), 14 age and gender-matched healthy controls (HC) and household contacts (HH) of DM group. The microbiota composition was analyzed by 16sRNA microbial profiling approach. RESULTS: Substantial differences were found in the richness of gut microbiota and the variation of bacteria population in DM compared to HC, and DN compared to DM, respectively. DM could be accurately distinguished from age/gender-matched healthy controls by the variable of genus g_Prevotella_9 (AUC = 0.9), and DN patients could be accurately distinguished from age/gender-matched DM by the variables of two genera (g_Escherichia-Shigella and g_Prevotella_9, AUC = 0.86). The microbiota composition of HH group was close to that of HC group, and was different from DM group. Under the same diet, DM could be more accurately detected by the same genus (g_Prevotella_9, AUC = 0.92). CONCLUSION: Gut microbiota composition was explored to be related to the occurrence of biopsy-proven DN from DM. DM could be distinguished from HC by detecting g_Prevotella_9 level in feces, while DN was different from DM by the variables of g_Escherichia-Shigella and g_Prevotella_9, which potentially contributed to the physiopathological diagnosis of DN from DM.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Diabetic Nephropathies/microbiology , Gastrointestinal Microbiome/physiology , Adult , Bacteria/genetics , Bacteria/isolation & purification , Biopsy , DNA, Bacterial/analysis , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/pathology , Diet , Feces/microbiology , Female , Humans , Kidney/physiopathology , Male , Middle Aged , Prevotella/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL