1
|
Kang Y, Kang X, Yang H, Liu H, Yang X, Liu Q, Tian H, Xue Y, Ren P, Kuang X, Cai Y, Tong M, Li L, Fan W. Lactobacillus acidophilus ameliorates obesity in mice through modulation of gut microbiota dysbiosis and intestinal permeability. Pharmacol Res 2022; 175:106020. [PMID: 34896249 DOI: 10.1016/j.phrs.2021.106020] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023]
Abstract
Obesity associated with low-grade chronic inflammation and intestinal dysbiosis is considered as a worldwide public health crisis. In the meanwhile, different probiotics have demonstrated beneficial effects on this condition, thus increasing the interest in the development of probiotic treatments. In this context, the aim of this study is to investigate the anti-obesity effects of potential probiotic Lactobacillus acidophilus isolated from the porcine gut. Then, it is found that L. acidophilus reduces body weight, fat mass, inflammation and insulin resistance in mice fed with a high-fat diet (HFD), accompanied by activation in brown adipose tissue (BAT) as well as improvements of energy, glucose and lipid metabolism. Besides, our data indicate that L. acidophilus not only reverses HFD-induced gut dysbiosis, as indicated by the decreased Firmicutes-to-Bacteroidetes ratios and endotoxin bearing Gram-negative bacteria levels, but also maintains intestinal barrier integrity, reduces metabolic endotoxemia, and inhibits the TLR4 / NF- κB signaling pathway. In addition, the results of microbiome phenotype prediction by BugBase and bacterial functional potential prediction using PICRUSt show that L. acidophilus treatment improves the gut microbiota functions involving metabolism, immune response, and pathopoiesia. Furthermore, the anti-obesity effect is transmissible via horizontal faeces transfer from L. acidophilus-treated mice to HFD-fed mice. According to our data, it is seen that L. acidophilus could be a good candidate for probiotic of ameliorating obesity and associated diseases such as hyperlipidemia, nonalcoholic fatty liver diseases, and insulin resistance through its anti-inflammatory properties and alleviation of endothelial dysfunction and gut dysbiosis.
Collapse
|
|
3 |
116 |
2
|
Kang Y, Li Y, Du Y, Guo L, Chen M, Huang X, Yang F, Hong J, Kong X. Konjaku flour reduces obesity in mice by modulating the composition of the gut microbiota. Int J Obes (Lond) 2019; 43:1631-1643. [PMID: 30242233 DOI: 10.1038/s41366-018-0187-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/23/2018] [Accepted: 07/02/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Changes in the intestinal flora composition is referred to as dysbiosis, which is related to obesity development, thus supporting the potential roles of nutrients acting on intestinal flora to exert salutary effects on energetic metabolism of host. Dietary fiber has been known to affect the composition of intestinal flora. The aim of the present study was to investigate the functional effects of konjac flour (KF) on obesity control in respect to improving inflammation, metabolism, and intestinal barrier function, and the possible association of the effects with intestinal flora composition changes. METHODS Mice (n = 30) were randomly divided into control group (n = 10), high-fat-diet (HFD) group (n = 10), and KF intervention group (n = 10), followed by feeding for 12 weeks and with adding a KF daily supplementation for the treatment group. Body weight, fat accumulation, inflammation, and energetic metabolism markers in multiple tissues and the gut microbiota of the mice were examined at the end of the experiment. RESULTS The KF supplementation significantly reduced the gains in weight, fat mass, as well as adipocyte size of HFD mice and lowered the serum TC, leptin (LEP), thiobarbituric acid-reacting substance (TBARS), IL-6, and lipopolysaccharide (LPS) levels in HFD mice. KF also upregulated the expression of intestinal mucosa protein gene Intection and tight junction ZO-1 in HFD mice, as well as upregulate the expression of energy metabolism genes PPARα and CPT-1 as well as the fat metabolism gene HLS in livers and fat tissues, and downregulate that of fat synthesis gene PPARγ (p < 0.05). The KF treatment increases the α-diversity and change the β-diversity of the intestinal microflora in HFD mice and boosted the abundances of some obesity-related beneficial microorganisms (such as Megasphaera elsdenii) in the intestinal microflora of HFD mice, while reduced those of harmful microorganisms (such as Alistipes, Alloprevotella, Bacteroides acidifaciens, and Parabacteroides goldsteinii). The abundance of Alistipes was positively correlated with weight, fat mass, serum TC, TG, LEP, IL-6, and LPS contents as well as PPARγ gene expression; while notably and negatively related to the expression of CPT-1 and HLS genes (p < 0.01). KF remarkably increased the abundance of Aerococcaceae, while reduced that of Alistipes finegoldii (p < 0.01). CONCLUSIONS Supplementation with KF achieves favorable effects on treating obesity, improving inflammatory response, metabolism, and intestinal barrier function, by regulating intestinal microfloral structure in HFD-fed mice.
Collapse
|
|
6 |
104 |
3
|
Kang Y, Cai Y. Gut microbiota and hypertension: From pathogenesis to new therapeutic strategies. Clin Res Hepatol Gastroenterol 2018; 42:110-117. [PMID: 29102544 DOI: 10.1016/j.clinre.2017.09.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 09/07/2017] [Accepted: 09/29/2017] [Indexed: 02/08/2023]
Abstract
Hypertension (HTN) has become a global public health concern and a major risk factor for cardiovascular, cerebrovascular, and kidney diseases. The complex interplay of genetic and environmental influences is important for the development of the disease. Accumulating evidence has illustrated the association of dysbiosis of gut microbiota with hypertension. Certain gut microbial strains may play either a pathogenic or a protective role in the development of hypertension. Oral probiotics can therefore represent a therapeutic approach for hypertension treatment. However, the relevant scientific work has only just begun, and the available data in this field remain limited. Fortunately, recent technological developments that permit identification of microbes and their products using culture-independent molecular detection techniques. In this review, we summarize the role of gut microbiota in hypertension progression, and probiotics in the treatment of hypertension.
Collapse
|
Review |
7 |
47 |
4
|
Liu Q, Tian H, Kang Y, Tian Y, Li L, Kang X, Yang H, Wang Y, Tian J, Zhang F, Tong M, Cai H, Fan W. Probiotics alleviate autoimmune hepatitis in mice through modulation of gut microbiota and intestinal permeability. J Nutr Biochem 2021; 98:108863. [PMID: 34517094 DOI: 10.1016/j.jnutbio.2021.108863] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 05/04/2021] [Accepted: 07/25/2021] [Indexed: 02/08/2023]
Abstract
Autoimmune hepatitis (AIH) is an immune-mediated type of chronic liver inflammation accompanied by intestinal flora imbalance. Probiotics have been reported to ameliorate imbalances in the intestinal flora. This study aimed to investigate the effects of compound probiotic in the AIH mouse model. AIH mice were gavaged with compound probiotic and injected intraperitoneally with dexamethasone (dex) for 42 days. The results showed that these treatments suppressed hepatic inflammatory cell infiltration, serum transaminase, and Th1 and Th17 cells. However, Treg cells were increased only in the probiotics group, which indicates an immunomodulatory role of the compound probiotic. The compound probiotic maintained intestinal barrier integrity, blocked lipopolysaccharide (LPS) translocation, and inhibited the activation of the TLR4/NF-κB pathway and the production of inflammatory factors in the liver and ileum. Moreover, the compound probiotic treatment increased the abundance of beneficial bacteria and reduced the abundance of potentially harmful bacteria in gut. Compound probiotic may improve ileal barrier function while increasing the diversity of the intestinal flora, blocking the translocation of gut-derived LPS to the liver and therefore preventing activation of the TLR4/NF-κB pathway. The resulting inhibition of pro-inflammatory factor production facilitates AIH remission.
Collapse
|
|
4 |
43 |
5
|
Kang YB, Cai Y, Zhang H. Gut microbiota and allergy/asthma: From pathogenesis to new therapeutic strategies. Allergol Immunopathol (Madr) 2017; 45:305-309. [PMID: 28029408 DOI: 10.1016/j.aller.2016.08.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/01/2016] [Indexed: 02/05/2023]
Abstract
Asthma and atopy, classically associated with hyper-activation of the T helper 2 (Th2) arm of adaptive immunity, are among the most common chronic illnesses worldwide. Emerging evidence relates atopy and asthma to the composition and function of gut microbiota composition. Moreover, certain gut microbial strains have been shown to inhibit or attenuate immune responses associated with chronic inflammation in experimental models. Although still a relatively nascent field of research, evidence to date suggests that the gut microbiome may represent fertile targets for prevention or management of allergic asthma and other diseases in which adaptive immune dysfunction is a prominent feature. The oral probiotics/prebiotic represents a possible therapeutic for improving asthma and allergic disease. Especially, recent technological developments that permit identification of microbes and their products using culture-independent molecular detection techniques. In this review, we literaturely summarise the aggravation or improvement of metabolic diseases by role of gut microbiota, probiotics/prebiotic treatment.
Collapse
|
Review |
8 |
43 |
6
|
Kang Y, Cai Y, Yang Y. The Gut Microbiome and Hepatocellular Carcinoma: Implications for Early Diagnostic Biomarkers and Novel Therapies. Liver Cancer 2022; 11:113-125. [PMID: 35634424 PMCID: PMC9109080 DOI: 10.1159/000521358] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/04/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks the third place among all causes inducing cancer-associated mortality, worldwide. HCC nearly exclusively occurs in cases suffering from chronic liver disease (CLD), which results from the vicious cycle of liver damage, inflammation, and regeneration possibly lasting for dozens of years. Recently, more and more investigation on microbiome-gut-liver axis enhances our understanding toward how gut microbiota promotes liver disease and even HCC development. In this review, we summarize the mechanisms underlying the effect of gut microbiota on promoting HCC occurrence, with the focus on key pathways such as bacterial dysbiosis, leaky gut, bacterial metabolites, and microorganism-related molecular patterns, which promote liver inflammation, genotoxicity, and fibrosis that finally lead to cancer occurrence. Furthermore, we discuss gut microbiota's important potential to be the early diagnostic biomarker for HCC. Gut microbiota may be the candidate targets to simultaneously prevent CLD and HCC occurrence among advanced liver disease cases. We outlook the gut microbiota-targeting treatments in detail to prevent CLD and HCC progression.
Collapse
|
Review |
3 |
36 |
7
|
Kang Y, Cai Y. Gut microbiota and hepatitis-B-virus-induced chronic liver disease: implications for faecal microbiota transplantation therapy. J Hosp Infect 2017; 96:342-348. [PMID: 28545829 DOI: 10.1016/j.jhin.2017.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/09/2017] [Indexed: 02/08/2023]
Abstract
Hepatitis B is one of the most common infectious diseases globally. It has been estimated that there are 350 million chronic hepatitis B virus (HBV) carriers worldwide. The liver is connected to the small intestine by the bile duct, which carries bile formed in the liver to the intestine. Nearly all of the blood that leaves the stomach and intestines must pass through the liver. Human intestines contain a wide diversity of microbes, collectively termed the 'gut microbiota'. Gut microbiota play a significant role in host metabolic processes and host immune modulation, and influence host development and physiology (organ development). Altered gut microbiota is a common complication in liver disease. Changes in intestinal microbiota seem to play an important role in induction and promotion of HBV-induced chronic liver disease progression, and specific species among the intestinal commensal bacteria may play either a pathogenic or a protective role in the development of HBV-induced chronic liver disease. Thus, the gut microbiome may represent fertile targets for prevention or management of HBV-induced chronic liver disease. Faecal microbiota transplantation (FMT) may be a useful therapy for HBV-related disease in the future. However, the data available in this field remain limited, and relevant scientific work has only just commenced. New technologies have enabled systematic studies of gut microbiota, and provided more realistic information about its composition and pathological variance. This review summarizes the cutting edge of research into the relationship between gut microbiota and HBV-induced chronic liver disease, and the future prospects of FMT therapy.
Collapse
|
Review |
8 |
34 |
8
|
Kang Y, Kuang X, Yan H, Ren P, Yang X, Liu H, Liu Q, Yang H, Kang X, Shen X, Tong M, Li L, Wang X, Guo L, Ma J, Zhang F, Fan W. A Novel Synbiotic Alleviates Autoimmune Hepatitis by Modulating the Gut Microbiota-Liver Axis and Inhibiting the Hepatic TLR4/NF-κB/NLRP3 Signaling Pathway. mSystems 2023; 8:e0112722. [PMID: 36794950 PMCID: PMC10134874 DOI: 10.1128/msystems.01127-22] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] [Imported: 11/18/2024] Open
Abstract
Autoimmune hepatitis (AIH) is a liver disease characterized by chronic liver inflammation. The intestinal barrier and microbiome play critical roles in AIH progression. AIH treatment remains challenging because first-line drugs have limited efficacy and many side effects. Thus, there is growing interest in developing synbiotic therapies. This study investigated the effects of a novel synbiotic in an AIH mouse model. We found that this synbiotic (Syn) ameliorated liver injury and improved liver function by reducing hepatic inflammation and pyroptosis. The Syn reversed gut dysbiosis, as indicated by an increase in beneficial bacteria (e.g., Rikenella and Alistipes) and a decrease in potentially harmful bacteria (e.g., Escherichia-Shigella) and lipopolysaccharide (LPS)-bearing Gram-negative bacterial levels. The Syn maintained intestinal barrier integrity, reduced LPS, and inhibited the TLR4/NF-κB and NLRP3/Caspase-1 signaling pathway. In addition, microbiome phenotype prediction by BugBase and bacterial functional potential prediction using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) showed that Syn improved gut microbiota function involving inflammatory injury, metabolism, immune response, and pathopoiesia. Furthermore, the new Syn was as effective as prednisone against AIH. Therefore, this novel Syn could be a candidate drug for alleviating AIH through its anti-inflammatory and antipyroptosis properties that relieve endothelial dysfunction and gut dysbiosis. IMPORTANCE Synbiotics can ameliorate liver injury and improve liver function by reducing hepatic inflammation and pyroptosis. Our data indicate that our new Syn not only reverses gut dysbiosis by increasing beneficial bacteria and decreasing lipopolysaccharide (LPS)-bearing Gram-negative bacteria but also maintains intestinal barrier integrity. Thus, its mechanism might be associated with modulating gut microbiota composition and intestinal barrier function by inhibiting the TLR4/NF-κB/NLRP3/pyroptosis signaling pathway in the liver. This Syn is as effective as prednisone in treating AIH without side effects. Based on these findings, this novel Syn represents a potential therapeutic agent for AIH in clinical practice.
Collapse
|
research-article |
2 |
31 |
9
|
Li M, Si S, Hao B, Zha Y, Wan C, Hong S, Kang Y, Jia J, Zhang J, Li M, Zhao C, Tu Y, Zhou S, Peng L. Mild alkali-pretreatment effectively extracts guaiacyl-rich lignin for high lignocellulose digestibility coupled with largely diminishing yeast fermentation inhibitors in Miscanthus. BIORESOURCE TECHNOLOGY 2014; 169:447-454. [PMID: 25079210 DOI: 10.1016/j.biortech.2014.07.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/03/2014] [Accepted: 07/05/2014] [Indexed: 05/03/2023] [Imported: 11/18/2024]
Abstract
In this study, various alkali-pretreated lignocellulose enzymatic hydrolyses were evaluated by using three standard pairs of Miscanthus accessions that showed three distinct monolignol (G, S, H) compositions. Mfl26 samples with elevated G-levels exhibited significantly increased hexose yields of up to 1.61-fold compared to paired samples derived from enzymatic hydrolysis, whereas Msa29 samples with high H-levels displayed increased hexose yields of only up to 1.32-fold. In contrast, Mfl30 samples with elevated S-levels showed reduced hexose yields compared to the paired sample of 0.89-0.98 folds at p<0.01. Notably, only the G-rich biomass samples exhibited complete enzymatic hydrolysis under 4% NaOH pretreatment. Furthermore, the G-rich samples showed more effective extraction of lignin-hemicellulose complexes than the S- and H-rich samples upon NaOH pretreatment, resulting in large removal of lignin inhibitors to yeast fermentation. Therefore, this study proposes an optimal approach for minor genetic lignin modification towards cost-effective biomass process in Miscanthus.
Collapse
|
|
11 |
26 |
10
|
Pan W, Kang Y. Gut microbiota and chronic kidney disease: implications for novel mechanistic insights and therapeutic strategies. Int Urol Nephrol 2018; 50:289-299. [PMID: 28849345 DOI: 10.1007/s11255-017-1689-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/23/2017] [Indexed: 02/05/2023]
Abstract
The complicated communities of microbiota colonizing the human gastrointestinal tract exert a strong function in health maintenance and disease prevention. Indeed, accumulating evidence has indicated that the intestinal microbiota plays a key role in the pathogenesis and development of chronic kidney disease (CKD). Modulation of the gut microbiome composition in CKD may contribute to the accumulation of gut-derived uremic toxins, high circulating level of lipopolysaccharides and immune deregulation, all of which play a critical role in the pathogenesis of CKD and CKD-associated complications. In this review, we discuss the recent findings on the potential impact of gut microbiota in CKD and the underlying mechanisms by which microbiota can influence kidney diseases and vice versa. Additionally, the potential efficacy of pre-, pro- and synbiotics in the restoration of healthy gut microbia is described in detail to provide future directions for research.
Collapse
|
Review |
7 |
22 |
11
|
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with progressive joint disorder. The complex interplay of genetic and environmental influences is important for the development of the disease. A growing body of evidence has shed light on the association of dysbiosis of gut microbiota with RA. Certain gut microbial strains have been shown to inhibit or attenuate immune responses in RA experimental models, suggesting that specific species among intestinal commensal bacteria may play either a pathogenic or a protective role in the development of RA. Oral intake of probiotics/prebiotics can therefore represent a therapeutic approach for RA treatment. However, the relevant scientific work has only just begun, and the available data in this field remain limited. Fortunately, utilization of new sequencing technologies allows expanded research on the association of intestinal bacterial flora and human diseases to be attempted. In this review, we summarize the role of gut microbiota in RA progression and address how specific bacterial strains regulate the immune response in disease process. Probiotics/prebiotics in the treatment of RA is also discussed.
Collapse
|
Review |
8 |
21 |
12
|
Kang Y, Cai Y. The development of probiotics therapy to obesity: a therapy that has gained considerable momentum. Hormones (Athens) 2018; 17:141-151. [PMID: 29858841 DOI: 10.1007/s42000-018-0003-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/06/2018] [Indexed: 02/06/2023]
Abstract
Obesity is a growing epidemic worldwide. The most frequent cause leading to the development of obesity is an imbalance between energy intake and energy expenditure. The gut microbiota is an environmental factor involved in obesity and metabolic disorders which reveals that obese animal and human subjects present alterations in the composition of the gut microbiota compared to their lean counterparts. Furthermore, evidence has so far demonstrated that the gut microbiota, which influences whole-body metabolism, by affecting energy balance, but also inflammation and gut barrier function, integrates peripheral and central food intake regulatory signals, thereby altering body weight. At the same time, these data suggest that species of intestinal commensal bacteria may play either a pathogenic or a protective role in the development of obesity. Though still a relatively nascent field of research, evidence to date suggests that manipulating the gut microbiome may represent effective treatment for the prevention or management of obesity. Various studies have described the beneficial effects of specific bacteria on the characteristics of obesity. However, the available data in this field remain limited and the relevant scientific work has only recently begun. This review aims to summarize the notable advances and contributions in the field that may prove useful for identifying probiotics that target obesity and its related disorders.
Collapse
|
Review |
7 |
21 |
13
|
Kang Y, Kang X, Zhang H, Liu Q, Yang H, Fan W. Gut Microbiota and Parkinson's Disease: Implications for Faecal Microbiota Transplantation Therapy. ASN Neuro 2021; 13:17590914211016217. [PMID: 34053243 PMCID: PMC8165863 DOI: 10.1177/17590914211016217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) ranks the second place among neurodegenerative diseases in terms of its morbidity, which affects 1-2% people aged over 65 years. In addition to genetics, some environmental factors may exert vital parts in PD occurrence as well. At present, more and more studies are conducted to elucidate the association between gut microbial dysbiosis and the incidence of PD. Gut microbial dysbiosis has a certain effect on both the central nervous system (CNS) and the enteric nervous system (ENS), which indicates that there is a gut-microbiota-brain axis that induces CNS disorders. Some gut microbial strains are suggested to suppress or weaken the neuroinflammation- and gut-inflammation-immune responses, which suggests the protective and pathogenic effects of certain gut microbial species on PD progression. Therefore, gut microbiome may contain plenty of targets for preventing and managing PD. Faecal microbiota transplantation (FMT) may serve as a direct and useful treatment for PD in the future. Nonetheless, there is little available scientific research in this field. The present work reviewed the latest research to examine the association of gut microbiota with PD, and the future prospects of FMT treatment.
Collapse
|
Review |
4 |
19 |
14
|
Kang Y, Cai Y. Altered Gut Microbiota in HIV Infection: Future Perspective of Fecal Microbiota Transplantation Therapy. AIDS Res Hum Retroviruses 2019; 35:229-235. [PMID: 29877092 DOI: 10.1089/aid.2017.0268] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
HIV infection progressively destroys CD4+ mononuclear cells, leading to profound cellular immune deficiency that manifests as life-threatening opportunistic infections and malignancies (i.e., AIDS). Gut microbiota plays key roles in the modulation of host metabolism and gene expression, maintenance of epithelial integrity, and mediation of inflammatory and immunity. Hence, the normal intestinal microbiota plays a major role in the maintenance of health and disease prevention. In fact, a large number of studies have shown that the alteration of the gut microbiota contributes to the pathogenesis of several diseases, such as inflammatory bowel diseases, irritable bowel syndrome, metabolic diseases, anorexia nervosa, autoimmune diseases, multiple sclerosis, cancer, neuropsychiatric disorders, and cardiovascular diseases. Recently, accumulating evidence has shed light on the association of dysbiosis of gut microbiota with HIV infection. Hence, the modification of gut microbiota may be a potential therapeutic tool. Fecal microbiota transplantation may improve the conditions of patients with HIV infection by manipulating the human intestinal bacteria. However, the relevant research is very limited, and a large amount of scientific research work needs to be done in the near future.
Collapse
|
Review |
6 |
18 |
15
|
Kang YB, Cai Y. Faecal microbiota transplantation enhances efficacy of immune checkpoint inhibitors therapy against cancer. World J Gastroenterol 2021; 27:5362-5375. [PMID: 34539138 PMCID: PMC8409158 DOI: 10.3748/wjg.v27.i32.5362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/03/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Even though immune checkpoint inhibitors (ICIs) are effective on multiple cancer types, there are still many non-responding patients. A possible factor put forward that may influence the efficacy of ICIs is the gut microbiota. Additionally, faecal microbiota transplantation may enhance efficacy of ICIs. Nevertheless, the data available in this field are insufficient, and relevant scientific work has just commenced. As a result, the current work reviewed the latest research on the association of gut microbiota with ICI treatments based on anti-programmed cell death protein 1 antibody and anti- cytotoxic T-lymphocyte-associated protein 4 antibody and explored the therapeutic potential of faecal microbiota transplantation in combination with ICI therapy in the future.
Collapse
|
Minireviews |
4 |
17 |
16
|
Kang Y, Cai Y. Gut microbiota and obesity: implications for fecal microbiota transplantation therapy. Hormones (Athens) 2017; 16:223-234. [PMID: 29278509 DOI: 10.14310/horm.2002.1742] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/14/2017] [Indexed: 02/05/2023]
Abstract
Obesity is a major public health issue as it is causally associated with several chronic disorders, including type-2 diabetes, cerebrovascular disease (CVD), and cancer. In the United States and other countries worldwide, the obesity epidemic has drastically impacted the status of health of millions as well as healthcare costs. Aside from poor diet, hygiene, and genetics, there are many other factors thought to play an important role in the emergence of obesity. Nowadays, accumulating evidence is elucidating the relation of dysbiosis of intestinal bacteria with obesity and metabolic disorders. Certain gut microbial strains have been shown to inhibit or attenuate immune responses related to chronic inflammation in experimental models, suggesting that specific species among gut microbiota may play either a protective or a pathogenic role in the progression of obesity. Fecal microbiota transplantation (FMT) can therefore represent a therapeutic approach for obesity treatment. FMT is a relatively straightforward therapy that manipulates the human gastrointestinal (GI) microbiota by transferring healthy donor microbiota into an existing but disturbed microbial ecosystem. However, the relevant scientific work is still in its early stages. In this review, we summarize the cutting-edge research being done into FMT treatment of obesity, current issues in FMT treatment, and the future of FMT and microbial therapeutics.
Collapse
|
Review |
8 |
15 |
17
|
Kang Y, Cai Y. Future prospect of faecal microbiota transplantation as a potential therapy in asthma. Allergol Immunopathol (Madr) 2018; 46:307-309. [PMID: 28803667 DOI: 10.1016/j.aller.2017.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 02/08/2023]
Abstract
There is convincing evidence from both human and animal studies suggesting that the gut microbiota plays an important role in regulating immune responses associated with the development of asthma. Certain intestinal microbial strains have been demonstrated to suppress or impair immune responsiveness in asthma experimental models, suggesting that specific species among gut commensal microbiota may play either a morbific or phylactic role in the progression of asthma. Evidence to date suggests that the intestinal microbiota represent fertile targets for prevention or management of asthma. The faecal microbiota transplantation (FMT) is a rather straightforward therapy that manipulates the human gastrointestinal (GI) microbiota, by which a healthy donor microbiota is transferred into an existing but disturbed microbial ecosystem. The FMT may therefore represent a therapeutic approach for asthma treatment in the foreseeable future. At present, FMT therapy for asthma is very limited and should be actively studied. Considerable efforts are needed to increase our knowledge in the field of FMT therapy for asthma. In this review, we aimed to provide several insights into the development of FMT therapy for asthma.
Collapse
|
Review |
7 |
15 |
18
|
Du Y, Neng Q, Li Y, Kang Y, Guo L, Huang X, Chen M, Yang F, Hong J, Zhou S, Zhao J, Yu F, Su H, Kong X. Gastrointestinal Autonomic Neuropathy Exacerbates Gut Microbiota Dysbiosis in Adult Patients With Type 2 Diabetes Mellitus. Front Cell Infect Microbiol 2022; 11:804733. [PMID: 35211420 PMCID: PMC8861497 DOI: 10.3389/fcimb.2021.804733] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/17/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The diabetic autonomic neuropathy is one of the most common complications in type 2 diabetes mellitus (T2DM), especially gastrointestinal autonomic neuropathy (GAN), which occurs in up to 75% of patients. The study aimed to investigate the gut microbiota composition, structure, and function in T2DM patients with GAN (T2DM_GAN) and set up a link between gut microbiota and clinical characteristics of patients. METHODS DNA was extracted from fecal samples of three groups using the kit method: healthy volunteers (n = 19), the patients with T2DM (n = 76), and T2DM_GAN (n = 27). Sequencing of 16S ribosomal DNA was performed using the MiSeq platform. RESULTS According to the clinical data, higher age, lower triglyceride, and lower body mass index were the main features of patients with T2DM_GAN. The gut microbiota analysis showed that Bacteroidetes, Firmicutes, and Proteobacteria constituted the three dominant phyla in healthy individuals. In addition, the gut microbiota structure and function of T2DM_GAN patients were clearly different from that of T2DM patients. T2DM patients were characterized by Fusobacteria, Fusobacteriia, Fusobacteriales, Fusobacteriaceae, Fusobacterium, Lachnoclostridium, and Fusobacterium_mortiferum. Those gut microbiota may be involved in carotenoid and flavonoid biosyntheses. Relatively, the Gammaproteobacteria, Enterobacteriales, Enterobacteriaceae, Escherichia-Shigella, Megasphaera, Escherichia_coli, and Megasphaera_elsdenii were characteristic in the T2DM_GAN patients. Those may be involved in bacterial invasion of epithelial cells and pathogenic Escherichia coli infection. CONCLUSIONS GAN exacerbated gut microbiota dysbiosis in adult patients with T2DM. The findings indicated that phyla Fusobacteria and class Gammaproteobacteria were closely related to the occurrence of T2DM. Especially the latter may promote T2DM_GAN.
Collapse
|
research-article |
3 |
13 |
19
|
Kang Y, Kang X, Cai Y. The gut microbiome as a target for adjuvant therapy in insomnia disorder. Clin Res Hepatol Gastroenterol 2022; 46:101834. [PMID: 34800683 DOI: 10.1016/j.clinre.2021.101834] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
Insomnia is a type of sleep disorder which has negative impacts on the quality of life, mood, cognitive function and health of humans. The etiology of insomnia may be related to many factors such as genetics, biochemistry, neuroendocrine, immune, and psychosocial factors. However, the detailed pathological aspects of insomnia remain unclear. Recent investigation of the microbiome-gut-brain axis enhances our understanding of the role of the gut microbiota in brain-related diseases. Gut microbiome has been shown to be associated with insomnia. However, the available data in this field remain limited and the relevant scientific work has only recently begun. This review aims to summarize the recent literature as an aid to better understanding how the alteration of gut microbiota composition contributes to insomnia while evaluating and prospecting the therapeutic effect of modulating gut microbiota in the treatment of insomnia based on previous publications.
Collapse
|
Review |
3 |
13 |
20
|
Liu H, Kang X, Yang X, Yang H, Kuang X, Ren P, Yan H, Shen X, Kang Y, Li L, Wang X, Guo L, Tong M, Fan W. Compound Probiotic Ameliorates Acute Alcoholic Liver Disease in Mice by Modulating Gut Microbiota and Maintaining Intestinal Barrier. Probiotics Antimicrob Proteins 2023; 15:185-201. [PMID: 36456838 DOI: 10.1007/s12602-022-10005-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 12/03/2022] [Imported: 11/18/2024]
Abstract
Alcoholic liver disease (ALD) is a worldwide health threaten lack of effective treatment. Gut dysbiosis and concomitant augmented intestinal permeability are strongly implicated in the pathogenesis and progression of ALD. Research on the protective effect of probiotics on ALD is limited, and more effective intestinal microecological regulators and the related mechanisms still need to be further explored. In the present study, the protective effects and mechanisms of a compound probiotic against acute alcohol-induced liver injury in vivo were explod. It was showed that the compound probiotic ameliorated liver injury in acute ALD mice and stabilized the levels of ALT, AST, and TG in serum. The compound probiotic reversed acute alcohol-induced gut dysbiosis and maintained the intestinal barrier integrity by upregulating the production of mucus and the expression of tight junction (TJ) proteins and thus reduced LPS level in liver. Meanwhile, the compound probiotic reduced inflammation level by inhibiting TLR4/NF-κB signaling pathway and suppressed oxidative stress level in liver. Furthermore, the compound probiotic alleviated liver lipid accumulation by regulating fatty acid metabolism-associated genes and AMPK-PPARα signaling pathway. Noteworthy, fecal microbiota transplantation (FMT) realized comparable protective effect with that of compound probiotic. In conclusion, present study demonstrates the beneficial effects and underlying mechanism of the compound probiotic against acute alcohol-induced liver injury. It provides clues for development of novel strategy for treatment of ALD.
Collapse
|
|
2 |
13 |
21
|
Kang Y, Zhang X, Cai Y, Su J, Kong X. Gut microbiota and metabolic disease: from pathogenesis to new therapeutic strategies. REVIEWS IN MEDICAL MICROBIOLOGY 2016; 27:141-152. [DOI: 10.1097/mrm.0000000000000085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gut microbiota is well known to regulate and maintain host metabolic and immune function. Change in stability and diversification of gut microbiota can affect progression of many metabolic diseases such as obesity, diabetes, liver disease, and so on. Studies on the association of the gut microbiota and host diseases are therefore significant, shedding light on the understanding of the role of gut microbiota in the development of such disease. In particular, human and animal model studies have explained how qualitative and quantitative alterations in the composition of gut microbiota are able to have an influence on the intestinal barrier, immune regulation, substance metabolism, nutrient absorption, energy distribution, toxin education, and so on. At the same time, these data suggest that species of intestinal commensal bacteria may play either a pathogenic or protective role in the development of metabolic diseases. The oral probiotic/prebiotic represents a possible therapeutic for improving metabolic diseases. However, the available data in this field remain limited, and the relevant scientific work has only just begun; especially, at present, new technologies have allowed the attempt at a systematic intestinal bacterial flora study, giving more realistic information about its composition and its pathological variance.In this review, we summarize the aggravation or improvement of metabolic diseases by the role of gut microbiota, and probiotic/prebiotic treatment with the help of available literature.
Collapse
|
|
9 |
11 |
22
|
Li Y, Kang Y, Du Y, Chen M, Guo L, Huang X, Li T, Chen S, Yang F, Yu F, Hong J, Kong X. Effects of Konjaku Flour on the Gut Microbiota of Obese Patients. Front Cell Infect Microbiol 2022; 12:771748. [PMID: 35300378 PMCID: PMC8921482 DOI: 10.3389/fcimb.2022.771748] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/28/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Gut microbiota have been thought to play a role in the emergence of obesity and metabolic disorders, thus dietary fiber may be an effective strategy for the management of obesity by modulating the gut microbiota. The aim of the present study was to investigate the effects of konjaku flour (KF) supplementation on treating obesity and regulating intestinal microbiota in obese adults. METHODS In a 5-week, randomized, double-blind, place-controlled trial, sixty-nine obese volunteers aged 25 to 35 with body mass index ≥28 kg/m2 were randomly assigned to receive KF or placebo (lotus root starch). Obesity index, blood parameters, and gut microbiota were analyzed. RESULTS KF remarkably reduced the body mass index (BMI), fat mass, percentage body fat (PBF), serum triglyceride (TG), glycated hemoglobin A1c (HbA1c), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) levels in the patients (p <0.05 or p <0.01). Meanwhile, high-throughput sequencing and bioinformatics analysis showed that the konjac flour treatment notably increased the α-diversity and changed the β-diversity of intestinal microflora in patients (p <0.01). Moreover, konjac flour could also evidently increase the abundance of some of the beneficial microorganisms related to obesity of patients, such as Lachnospiraceae, Roseburia, Solobacterium, R. inulinivorans, Clostridium perfringens, and Intestinimonas butyriciproducens, and reduce the abundance of the harmful microorganisms, such as Lactococcus, Bacteroides fragilis, Lactococcus garvieae, B. coprophilus, B. ovatus, and B. thetaiotaomicron (p <0.01). Specifically, C. perfringens was significantly negatively correlated with serum total cholesterol (TC) (p <0.01). CONCLUSION These results suggested that KF can achieve positive effects on treating obesity, which manifest on reducing BMI, fat mass, blood glucose, and blood lipid, improving hepatic function, and also regulating intestinal microfloral structure. Therefore, changes in gut microbiota may explain in part the effects of KF.
Collapse
|
Randomized Controlled Trial |
3 |
10 |
23
|
Kang Y, Cai Y, Pan W. Change in gut microbiota for eczema: Implications for novel therapeutic strategies. Allergol Immunopathol (Madr) 2018; 46:281-290. [PMID: 29279259 DOI: 10.1016/j.aller.2017.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/19/2017] [Indexed: 02/08/2023]
Abstract
Eczema is one of the most common inflammatory diseases, often constituting a lifelong burden for afflicted individuals. The complex interaction of host genetic and multiple environmental factors contribute to its pathogenesis. A relationship between maladjustment of gut microbiota and eczema has been brought into the light of day in most previous studies. In eczema preclinical models, specific intestinal microbial species have been demonstrated to prohibit or dwindle immune responsiveness, indicating that these strains among commensal gut bacteria may exert either a morbific or phylactic function in eczema progression. As such, oral probiotics can serve as a medicinal approach for eczema therapy. Given that relative scientific work is still at the early stage, only limited data are available in the field. New sequencing techniques have been fortunately performed to gain access to an extended research on the relationship between gut bacterial flora and human diseases. In the current review, we identified the role of intestinal microbiota in the development of eczema and how specific bacterial strains adjust the immune responsiveness in the midst of disease progression. Probiotics as an applicable treatment for eczema were evaluated in other threads as well.
Collapse
|
Review |
7 |
9 |
24
|
Kang Y, Pan W, Cai Y. Gut microbiota and colorectal cancer: insights into pathogenesis for novel therapeutic strategies. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2017; 55:872-880. [PMID: 29186641 DOI: 10.1055/s-0043-116387] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC), as a leading cause of cancer-related death, is triggered by the complex interplay of host genetics and environmental factors. Mounting evidence has shed light on the association of the gut microbiota dysbiosis with CRC. In CRC experimental models, certain gut microbial strains have been shown to inhibit or attenuate immune responses, indicating that specific species among intestinal commensal bacteria may play either a pathogenic or a protective role in the development of CRC. Oral intake of probiotics/prebiotics can therefore serve as a therapeutic approach for CRC treatment. Microbiota studies in cancer, however, are still at the early stage, lack of quantitative data for clinic application. Fortunately, sequencing-based technologies are a boon to further investigation on the association of the intestinal bacterial flora and human diseases. This review considers the evidence for the role of the gut microbiota in CRC development and progression, responsiveness to immune system, and the related therapeutic applications of probiotics/prebiotics.
Collapse
|
Review |
8 |
7 |
25
|
Kang Y, Cai Y. Epidemiology and Genetic Diversity of Rotavirus in Kunming, China, in 2015. Intervirology 2018; 61:9-13. [PMID: 29975941 DOI: 10.1159/000489309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/16/2018] [Indexed: 02/05/2023] Open
Abstract
Group A rotavirus (RVA) is a serious public problem in China, with a prevalence of 29.7% in diarrhea cases under 5 years of age. A total of 1,224 fecal specimens were collected in 2015 from children under 5 years of age with acute gastroenteritis in Kunming, China, for detection and characterization of rotavirus. The prevalence of RVA was 28.5%. The frequency of RVA detection was greatest (52%) among children aged 7-12 months. The following strains were the most common types: G9P(8) (58.2%), G3P(8) (14.9%), and G1P(8) (6.9%). The following strains were uncommon types: G3P(4), G8P(9), and G9P(4) (1.1%); G1P(4), G2P(8), and G4P(8) (0.9%); G2P(6), G3P(9), G4P(6), and G9P(6) (0.6%); and G4P(4) (0.3%). G3G1P(8) (5.2%), G1G2P(4) (0.9%), G1G9P(8) (0.6%), and G2P(4)P(8) (0.3%) were mixed types. This study documents the molecular epidemiology, genetics, and diversity of rotavirus strains in children under 5 years of age in Kunming, China, and suggests that it may be important to offer RVA vaccination and scientific evidence to stop and control RVA-related diarrhea.
Collapse
|
|
7 |
5 |