1
|
Dominique GM, Hammond C, Stack MS. The Gut Microbiome in Aging and Ovarian Cancer. AGING AND CANCER 2024; 5:14-34. [PMID: 39132604 PMCID: PMC11309124 DOI: 10.1002/aac2.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/30/2024] [Indexed: 08/13/2024]
Abstract
The gut microbiome changes with age and affects regions beyond the gut, including the ovarian cancer tumor microenvironment. In this review summarizing the literature on the gut microbiome in ovarian cancer and in aging, we note trends in the microbiota composition common to both phenomena and trends that are distinctly opposite. Both ovarian cancer and aging are characterized by an increase in proinflammatory bacterial species, particularly those belonging to phylum Proteobacteria and genus Escherichia, and a decrease in short chain fatty acid producers, particularly those in Clostridium cluster XIVa (family Lachnospiraceae) and the Actinobacteria genus Bifidobacterium. However, while beneficial bacteria from family Porphyromonadaceae and genus Akkermansia tend to increase with normal, healthy aging, these bacteria tend to decrease in ovarian cancer, similar to what is observed in obesity or unhealthy aging. We also note a lack in the current literature of research demonstrating causal relationships between the gut microbiome and ovarian cancer outcomes and research on the gut microbiome in ovarian cancer in the context of aging, both of which could lead to improvements to ovarian cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Gena M Dominique
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| | | | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
2
|
Wang K, Duan F, Sun T, Zhang Y, Lu L. Galactooligosaccharides: Synthesis, metabolism, bioactivities and food applications. Crit Rev Food Sci Nutr 2024; 64:6160-6176. [PMID: 36632761 DOI: 10.1080/10408398.2022.2164244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Prebiotics are non-digestible ingredients that exert significant health-promoting effects on hosts. Galactooligosaccharides (GOS) have remarkable prebiotic effects and structural similarity to human milk oligosaccharides. They generally comprise two to eight sugar units, including galactose and glucose, which are synthesized from substrate lactose by microbial β-galactosidase. Enzyme sources from probiotics have received particular interest because of their safety and potential to synthesize specific structures that are particularly metabolized by intestinal probiotics. Owing to advancements in modern analytical techniques, many GOS structures have been identified, which vary in degree of polymerization, glycosidic linkage, and branch location. After intake, GOS adjust gut microbiota which produce short chain fatty acids, and exhibit excellent biological activities. They selectively stimulate the proliferation of probiotics, inhibit the growth and adhesion of pathogenic bacteria, alleviate gastrointestinal, neurological, metabolic and allergic diseases, modulate metabolites production, and adjust ion storage and absorption. Additionally, GOS are safe and stable, with high solubility and clean taste, and thus are widely used as food additives. GOS can improve the appearance, flavor, taste, texture, viscosity, rheological properties, shelf life, and health benefits of food products. This review systemically covers GOS synthesis, structure identifications, metabolism mechanisms, prebiotic bioactivities and wide applications, focusing on recent advances.
Collapse
Affiliation(s)
- Ke Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feiyu Duan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Sun
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lili Lu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Afsar B, Afsar RE, Lentine KL. The impact of sodium-glucose cotransporter inhibitors on gut microbiota: a scoping review. J Diabetes Metab Disord 2024; 23:497-508. [PMID: 38932911 PMCID: PMC11196485 DOI: 10.1007/s40200-024-01435-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/10/2024] [Indexed: 06/28/2024]
Abstract
Studies consistently showed that sodium-glucose cotransporter inhibitors (SGLTi) have cardiovascular and renal benefits, independent of their glucose lowering effects. Recent studies showed that SGLTi might influence gut microbiota. We performed a narrative review of publications focusing on use of SGLTi and changes in gut microbiota. Most studies showed that use of SGLTi re-shapes gut microbiota. These studies are heterogeneous regarding in study designs, doses and types of drugs used (SGLT1i vs. SGLT2i, or SGLT1/2i in combination) and the methods used to determine gut microbiota. However, existing data showed that SGLTi might alter food fermentation and gut permeability, which might translate into clinical outcomes. Thus the objective of this review is to summarize and discuss the updated data regarding SGLTi and changes in gut microbiota for the first time and suggest further study points that needs to be discovered. Graphical Abstract
Collapse
Affiliation(s)
- Baris Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey
| | - Rengin Elsurer Afsar
- Suleyman Demirel University, School of Medicine, Department of Nephrology, Isparta, Turkey
- Saint Louis University, School of Medicine, Division of Nephrology, St. Louis, MO USA
| | - Krista L. Lentine
- Saint Louis University, School of Medicine, Division of Nephrology, St. Louis, MO USA
| |
Collapse
|
4
|
Bevilacqua A, Campaniello D, Speranza B, Racioppo A, Sinigaglia M, Corbo MR. An Update on Prebiotics and on Their Health Effects. Foods 2024; 13:446. [PMID: 38338581 PMCID: PMC10855651 DOI: 10.3390/foods13030446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Prebiotic compounds were originally defined as "a nondigestible food ingredient that beneficially affects the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the colon, and thus improves host health"; however, a significant modulation of the definition was carried out in the consensus panel of The International Scientific Association for Probiotics and Prebiotics (ISAPP), and the last definition states that "prebiotics are substrates that are selectively utilized by host microorganisms conferring a health benefit". Health effects of prebiotics compounds attracted the interest of researchers, food companies and Regulatory Agencies, as inferred by the number of articles on Scopus for the keywords "prebiotic" and "health effects", that is ca. 2000, for the period January 2021-January 2024. Therefore, the aim of this paper is to contribute to the debate on these topics by offering an overview of existing knowledge and advances in this field. A literature search was performed for the period 2012-2023 and after the selection of the most relevant items, the attention was focused on seven conditions for which at least 8-10 different studies were found, namely colorectal cancer, neurological or psychiatric conditions, intestinal diseases, obesity, diabetes, metabolic syndrome, and immune system disorders. In addition, the analysis of the most recent articles through the software VosViewer version 1.6.20 pointed out the existence of five clusters or macro-categories, namely: (i) pathologies; (ii) metabolic condvitions; (iii) structure and use in food; (iv) immunomodulation; (v) effect on gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Rosaria Corbo
- Department of the Science of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (A.B.); (D.C.); (B.S.); (A.R.); (M.S.)
| |
Collapse
|
5
|
Abe T. [Therapy for CKD and DKD]. Nihon Yakurigaku Zasshi 2023; 158:319-325. [PMID: 37394553 DOI: 10.1254/fpj.22133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Diabetic kidney disease is a major cause of renal failure that urgently necessitates a breakthrough in disease management. Specific remedies are needed for preventing Type 2 diabetes which causes significant changes in an array of plasma metabolites. By untargeted metabolome analysis, phenyl sulfate (PS) increased with the progression of diabetes. In experimental diabetes models, PS administration induces albuminuria and podocyte damage due to the mitochondrial dysfunction. By clinical diabetic kidney disease (DKD) cohort analysis, it was also confirmed that the PS levels significantly correlate with basal and predicted 2-year progression of albuminuria. Phenol is synthesized from dietary tyrosine by gut bacterial-specific tyrosine phenol-lyase (TPL), and absorbed phenol is metabolized into PS in the liver. Inhibition of TPL reduces not only the circulating PS level but also albuminuria in diabetic mice. TPL inhibitor did not significantly alter the major composition, showing the non-lethal inhibition of microbial-specific enzymes has a therapeutic advantage, with lower selective pressure for the development of drug resistance. Clinically, 362 patients in a multi-center clinical study in diabetic nephropathy cohort (U-CARE) were analyzed with full data. The basal plasma PS level significantly correlated with ACR, eGFR, age, duration, HbA1c and uric acid, but not with suPAR. Multiple regression analysis revealed that ACR was the only factor that significantly correlated with PS. By stratified logistic regression analysis, in the microalbuminuria group, PS was the only factor related to the amount of change in the 2-year ACR in all models. PS is not only an early diagnosis marker, but also a modifiable cause and therefore a target for the treatment of DKD. Reduction of microbiota-derived phenol by the inhibitor should represent another aspect for developing drugs of DKD prevention.
Collapse
Affiliation(s)
- Takaaki Abe
- Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine
- AMED Moon Program Manager
| |
Collapse
|
6
|
Liu X, Zhang M, Wang X, Liu P, Wang L, Li Y, Wang X, Ren F. Fecal microbiota transplantation restores normal fecal composition and delays malignant development of mild chronic kidney disease in rats. Front Microbiol 2022; 13:1037257. [PMID: 36532422 PMCID: PMC9748282 DOI: 10.3389/fmicb.2022.1037257] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/15/2022] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is associated with gut microbiome dysbiosis, but the role of intestinal flora in CKD treatment remains to be elucidated. Fecal microbiota transplantation (FMT) can be utilized to re-establish healthy gut microbiota for a variety of diseases, which offers new insight for treating CKD. First, 5/6 nephrectomy rats (Donor CKD) and sham rats (Donor Sham) were used as donors for FMT, and fecal metagenome were analyzed to explore potential therapeutic targets. Then, to assess the effect of FMT on CKD, sterilized 1/2 nephrectomy rats were transplanted with fecal microbiota from Donor sham (CKD/Sham) or Donor CKD (CKD/CKD) rats, and 1/2 nephrectomy rats without FMT (CKD) or no nephrectomy (Sham) were used as model control or normal control. Results showed that Bacteroides uniformis and Anaerotruncus sp. 1XD22-93 were enriched in Donor CKD, while Lactobacillus johnsonii and Lactobacillus intestinalis were reduced. In addition, the increased abundance of microbial functions included tryptophan metabolism and lysine degradation contributing to the accumulation of protein-bound uremic toxins (PBUTs) in Donor CKD. Genome analysis indicated that FMT successfully differentiated groups of gut microbes and altered specific gut microbiota after 1 week of treatment, with Bacteroides uniformis and Anaerotruncus sp. 1XD22-93 increasing in CKD/CKD group as well as Lactobacillus johnsonii and Lactobacillus intestinalis being improved in CKD/Sham group. In comparison to CKD group, substantial PBUT buildup and renal damage were observed in CKD/CKD. Interestingly, compared to CKD or CKD/CKD group, tryptophan metabolism and lysine degradation were efficiently suppressed in CKD/Sham group, while lysine biosynthesis was promoted. Therefore, FMT considerably reduced PBUTs accumulation. After FMT, PBUTs and renal function in CKD/Sham rats remained the same as in Sham group throughout the experimental period. In summary, FMT could delay the malignant development of CKD by modifying microbial amino acid metabolism through altering the microenvironment of intestinal flora, thereby providing a novel potential approach for treating CKD.
Collapse
Affiliation(s)
- Xiaoxue Liu
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ming Zhang
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China
| | - Xifan Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Ping Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Longjiao Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xiaoyu Wang
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Co-Constructed by Ministry of Education and Beijing Municipality, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Food Laboratory of Zhongyuan, Luohe, Henan, China
| |
Collapse
|
7
|
Shi J, Wang F, Tang L, Li Z, Yu M, Bai Y, Weng Z, Sheng M, He W, Chen Y. Akkermansia muciniphila attenuates LPS-induced acute kidney injury by inhibiting TLR4/NF-κB pathway. FEMS Microbiol Lett 2022; 369:6824436. [PMID: 36368696 DOI: 10.1093/femsle/fnac103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/25/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
Acute kidney injury (AKI) is a global public health hazard with high morbidity and mortality. Sepsis accounts for nearly half of all causes of AKI. Scientists have made a great effort to explore effective therapeutic agents with limited side effects in the treatment of AKI, but have had little success. With the development of gut flora study, Akkermansia muciniphila (A. muciniphila) has been proven to prevent different organs by regulating the inflammatory response. However, the reno-protective function is still unknown. Here, the AKI model was induced using lipopolysaccharide (LPS) in mice with or without pretreatment of A. muciniphila. Renal function and histological change were measured. Inflammatory factors were detected by ELISA and rt-PCR. TLR4/NF-κB signaling factors and NLRP3 inflammasome were tested by western blot and immunohistochemistry. Pretreatment of A. muciniphila markedly inhibited inflammatory response and ameliorated kidney histopathological changes. Furthermore, the TLR4, p-NF-κB p65, and downstream IκBα were notably activated in the model group and inhibited by A. muciniphila. A similar effect was found in the regulation of NLRP3 inflammasome. In conclusion, pretreatment with A. muciniphila could protect against LPS-induced AKI by inhibition of the TLR4/NF-κB pathway and NLRP3 inflammasome activation. It may be a new therapeutic strategy for AKI prevention and treatment in the future.
Collapse
Affiliation(s)
- Jun Shi
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Feng Wang
- Department of Analytical & Testing Center, Nanjing Medical University, Nanjing 211166, PR China
| | - Lei Tang
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Zhiqiang Li
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Manshu Yu
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Yu Bai
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zebin Weng
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Meixiao Sheng
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Weiming He
- Renal Division, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| | - Yugen Chen
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China
| |
Collapse
|
8
|
Cai Y, Gilbert MS, Gerrits WJ, Folkerts G, Braber S. Galacto-oligosaccharides alleviate lung inflammation by inhibiting NLRP3 inflammasome activation in vivo and in vitro. J Adv Res 2021; 39:305-318. [PMID: 35777914 PMCID: PMC9263649 DOI: 10.1016/j.jare.2021.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/29/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022] Open
Abstract
GOS suppress both local and systemic inflammation in lung infections. GOS reduce the M. haemolytica positivity in calves with lung infections. GOS inhibit NLRP3 inflammasome activation in vivo and in vitro. GOS decrease ATP production in PBECs induced by M. haemolytica. Direct anti-oxidative effects of GOS on lung cells are involved.
Introduction Objectives Methods Results Conclusion
Collapse
|
9
|
Melekoglu E, Samur FG. Dietary strategies for gut-derived protein-bound uremic toxins and cardio-metabolic risk factors in chronic kidney disease: A focus on dietary fibers. Crit Rev Food Sci Nutr 2021:1-15. [PMID: 34704501 DOI: 10.1080/10408398.2021.1996331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Chronic kidney disease (CKD) is associated with altered composition and function of gut microbiota. The cause of gut dysbiosis in CKD is multifactorial and encompasses the following: uremic state, metabolic acidosis, slow colonic transit, dietary restrictions of plant-based fiber-rich foods, and pharmacological therapies. Dietary restriction of potassium-rich fruits and vegetables, which are common sources of fermentable dietary fibers, inhibits the conversion of dietary fibers to short-chain fatty acids (SCFA), which are the primary nutrient source for the symbiotic gut microbiota. Reduced consumption of fermentable dietary fibers limits the population of SCFA-forming bacteria and causes dysbiosis of gut microbiota. Gut dysbiosis induces colonic fermentation of protein and formation of gut-derived uremic toxins. In this review, we discuss the roles and benefits of dietary fiber on gut-derived protein-bound uremic toxins and plant-based dietary patterns that could be recommended to decrease uremic toxin formation in CKD patients. Recent studies have indicated that dietary fiber supplementation may be useful to decrease gut-derived uremic toxin formation and slow CKD progression. However, research on associations between adherence of healthy dietary patterns and gut-derived uremic toxins formation in patients with CKD is lacking.
Collapse
Affiliation(s)
- Ebru Melekoglu
- Faculty of Health Sciences, Nutrition and Dietetics Department, Hacettepe University, Ankara, Turkey.,Faculty of Health Sciences, Nutrition and Dietetics Department, Cukurova University, Adana, Turkey
| | - F Gulhan Samur
- Faculty of Health Sciences, Nutrition and Dietetics Department, Hacettepe University, Ankara, Turkey
| |
Collapse
|
10
|
Melekoglu E, Cetinkaya MA, Kepekci-Tekkeli SE, Kul O, Samur G. Effects of prebiotic oligofructose-enriched inulin on gut-derived uremic toxins and disease progression in rats with adenine-induced chronic kidney disease. PLoS One 2021; 16:e0258145. [PMID: 34614017 PMCID: PMC8494360 DOI: 10.1371/journal.pone.0258145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/21/2021] [Indexed: 11/18/2022] Open
Abstract
Recent studies suggest that dysbiosis in chronic kidney disease (CKD) increases gut-derived uremic toxins (GDUT) generation, leads to systemic inflammation, reactive oxygen species generation, and poor prognosis. This study aimed to investigate the effect of oligofructose-enriched inulin supplementation on GDUT levels, inflammatory and antioxidant parameters, renal damage, and intestinal barrier function in adenine-induced CKD rats. Male Sprague-Dawley rats were divided into control group (CTL, n = 12) fed with standard diet; and CKD group (n = 16) given adenine (200 mg/kg/day) by oral gavage for 3-weeks to induce CKD. At the 4th week, CKD rats were subdivided into prebiotic supplementation (5g/kg/day) for four consecutive weeks (CKD-Pre, n = 8). Also, the control group was subdivided into two subgroups; prebiotic supplemented (CTL-Pre, n = 6) and non-supplemented group (CTL, n = 6). Results showed that prebiotic oligofructose-enriched inulin supplementation did not significantly reduce serum indoxyl sulfate (IS) but did significantly reduce serum p-Cresyl sulfate (PCS) (p = 0.002) in CKD rats. Prebiotic supplementation also reduced serum urea (p = 0.008) and interleukin (IL)-6 levels (p = 0.001), ameliorated renal injury, and enhanced antioxidant enzyme activity of glutathione peroxidase (GPx) (p = 0.002) and superoxide dismutase (SOD) (p = 0.001) in renal tissues of CKD rats. No significant changes were observed in colonic epithelial tight junction proteins claudin-1 and occludin in the CKD-Pre group. In adenine-induced CKD rats, oligofructose-enriched inulin supplementation resulted in a reduction in serum urea and PCS levels, enhancement of the antioxidant activity in the renal tissues, and retardation of the disease progression.
Collapse
Affiliation(s)
- Ebru Melekoglu
- Nutrition and Dietetics Department, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| | - M Alper Cetinkaya
- Laboratory Animals Application and Research Center, Hacettepe University, Ankara, Turkey
| | - S Evrim Kepekci-Tekkeli
- Department of Analytical Chemistry, Faculty of Pharmacy, Bezmialem Vakıf University, Istanbul, Turkey
| | - Oguz Kul
- Department of Pathology, Faculty of Veterinary Medicine, Kirikkale University, Kirikkale, Turkey
| | - Gulhan Samur
- Nutrition and Dietetics Department, Faculty of Health Sciences, Hacettepe University, Ankara, Turkey
| |
Collapse
|
11
|
Huang H, Li K, Lee Y, Chen M. Preventive Effects of Lactobacillus Mixture against Chronic Kidney Disease Progression through Enhancement of Beneficial Bacteria and Downregulation of Gut-Derived Uremic Toxins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7353-7366. [PMID: 34170659 DOI: 10.1021/acs.jafc.1c01547] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Gut dysbiosis is a major contributor to adverse chronic kidney disease (CKD) progression, and microbiota-based strategies could be considered as a novel therapeutic and preventative target. In this study, a probiotic screening platform based on gut-derived uremic toxin-reducing probiotics was developed and the underlying mechanism was further verified through a 0.2% adenine-induced CKD mouse model. Two strains (Lactobacillus paracasei and Lactobacillus plantarum) were selected due to their high clearance ability and named Lactobacillus mix (Lm). The results showed that Lm significantly improved the kidney function by reducing kidney injury and fibrotic-related proteins. Furthermore, Lm decreased oxidative stress and proinflammatory reactions and elevated immune responses in the kidney. Importantly, Lm reversed gut dysbiosis and restored the abundance of commensal bacteria, especially short-chain fatty acid producers, leading to improved intestinal barrier integrity via modulation of microbial composition and metabolite production. Taken together, these findings provided evidence that Lm could be a preventive approach against CKD.
Collapse
Affiliation(s)
- Hsiaowen Huang
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Section 3, Keelung Road, Taipei 10673, Taiwan
| | - Kuanyi Li
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Section 3, Keelung Road, Taipei 10673, Taiwan
| | - Yajane Lee
- Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, No. 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
- Department of Internal Medicine, National Taiwan University Veterinary Hospital, No. 153, Section 3, Keelung Road, Taipei 10672, Taiwan
| | - Mingju Chen
- Department of Animal Science and Technology, National Taiwan University, No. 50, Lane 155, Section 3, Keelung Road, Taipei 10673, Taiwan
- Center for Biotechnology, National Taiwan University, No. 81, Changxing Street, Taipei 10672, Taiwan
| |
Collapse
|
12
|
Thrombolome and Its Emerging Role in Chronic Kidney Diseases. Toxins (Basel) 2021; 13:toxins13030223. [PMID: 33803899 PMCID: PMC8003125 DOI: 10.3390/toxins13030223] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/25/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at an increased risk of thromboembolic complications, including myocardial infarction, stroke, deep vein thrombosis, and pulmonary embolism. These complications lead to increased mortality. Evidence points to the key role of CKD-associated dysbiosis and its effect via the generation of gut microbial metabolites in inducing the prothrombotic phenotype. This phenomenon is known as thrombolome, a panel of intestinal bacteria-derived uremic toxins that enhance thrombosis via increased tissue factor expression, platelet hyperactivity, microparticles release, and endothelial dysfunction. This review discusses the role of uremic toxins derived from gut-microbiota metabolism of dietary tryptophan (indoxyl sulfate (IS), indole-3-acetic acid (IAA), kynurenine (KYN)), phenylalanine/tyrosine (p-cresol sulfate (PCS), p-cresol glucuronide (PCG), phenylacetylglutamine (PAGln)) and choline/phosphatidylcholine (trimethylamine N-oxide (TMAO)) in spontaneously induced thrombosis. The increase in the generation of gut microbial uremic toxins, the activation of aryl hydrocarbon (AhRs) and platelet adrenergic (ARs) receptors, and the nuclear factor kappa B (NF-κB) signaling pathway can serve as potential targets during the prevention of thromboembolic events. They can also help create a new therapeutic approach in the CKD population.
Collapse
|
13
|
Armani RG, Carvalho AB, Ramos CI, Hong V, Bortolotto LA, Cassiolato JL, Oliveira NF, Cieslarova Z, do Lago CL, Klassen A, Cuppari L, Raj DS, Canziani MEF. Effect of fructooligosaccharide on endothelial function in CKD patients: a randomized controlled trial. Nephrol Dial Transplant 2021; 37:85-91. [PMID: 33411910 DOI: 10.1093/ndt/gfaa335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Microbiota-derived uremic toxins have been associated with inflammation that could corroborate with endothelial dysfunction (ED) and increase cardiovascular risk in patients with chronic kidney disease (CKD). This trial aimed to evaluate the effect of the prebiotic fructooligosaccharide (FOS) on endothelial function and arterial stiffness in nondialysis CKD patients. METHODS In a double-blind controlled trial, 46 nondiabetic CKD patients were randomized to receive 12 g/day of FOS or placebo (maltodextrin) for 3 months. Total p-cresyl sulfate (PCS) and indoxyl sulfate by high-performance liquid chromatography, urinary trimethylamine N-oxide by mass spectrometry, C-reactive protein, interleukin-6 (IL-6), serum nitric oxide and stroma-derived factor-1 alfa were measured at baseline and at the end of follow-up; endothelial function was assessed through flow-mediated dilatation (FMD) and arterial stiffness by pulse wave velocity (PWV). RESULTS The mean (± standard deviation) age of the study participants was 57.6 ± 14.4 years, with an estimated glomerular filtration rate of 21.3 ± 7.3 mL/min/1.73 m2. During the follow-up, regarding the inflammatory markers and uremic toxins, there was a significant decrease in IL-6 levels (3.4 ± 2.1 pg/mL versus 2.6 ± 1.4 pg/mL; P = 0.04) and a trend toward PCS reduction (55.4 ± 38.1 mg/L versus 43.1 ± 32.4 mg/L, P = 0.07) only in the prebiotic group. Comparing both groups, there was no difference in FMD and PWV. In an exploratory analysis, including a less severe ED group of patients (FMD ≥2.2% at baseline), FMD remained stable in the prebiotic group, while it decreased in the placebo group (group effect P = 0.135; time effect P = 0.012; interaction P = 0.002). CONCLUSIONS The prebiotic FOS lowered circulating levels of IL-6 in CKD patients and preserved endothelial function only in those with less damaged endothelium. No effect of FOS in arterial stiffness was observed.
Collapse
Affiliation(s)
- Rachel G Armani
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Aluizio B Carvalho
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Christiane I Ramos
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Valeria Hong
- Heart Institute, University of São Paulo, São Paulo, Brazil
| | | | | | - Natacha F Oliveira
- Department of Chemistry, Federal University of São Paulo, Diadema, Brazil
| | | | | | - Aline Klassen
- Department of Chemistry, Federal University of São Paulo, Diadema, Brazil
| | - Lilian Cuppari
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Maria Eugênia F Canziani
- Department of Medicine, Division of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Rui-Zhi T, Hui D, Jian-Chun L, Xia Z, Xiao-Jia W, Dan W, Jun-Ming F, Li W. Astragalus mongholicus Bunge and Panax Notoginseng Formula (A&P) Combined With Bifidobacterium Contribute a Renoprotective Effect in Chronic Kidney Disease Through Inhibiting Macrophage Inflammatory Response in Kidney and Intestine. Front Physiol 2020; 11:583668. [PMID: 33329031 PMCID: PMC7729014 DOI: 10.3389/fphys.2020.583668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022] Open
Abstract
There is increasing evidence that Chronic Kidney Disease (CKD) can cause intestinal dysfunction, which in turn aggravates the progression of kidney disease. Studies have shown that the immune response of macrophage plays an important role in promoting inflammation in kidney and intestine of CKD. Astragalus mongholicus Bunge and Panax notoginseng formula (A&P) is a widely used traditional medicine for the treatment of CKD in China, however, the underlying mechanism is largely unclear. In this study, we aimed to explore the role of A&P and Bifidobacterium combination treatment in regulation of inflammatory response of macrophage in kidney and intestine of CKD mouse, as well as the potential molecular mechanism. We established a CKD mouse model with 5/6 nephrectomy and a macrophage inflammatory cellular model with LPS and urotoxin in vivo and in vitro. The results showed that A&P combined with Bifidobacterium significantly reduced the expression and secretion of IL-1β, IL-6, TNFα, and MCP-1 in kidney and blood, as well as in inflammatory macrophage. Interestingly, A&P combined with Bifidobacterium strongly improved the intestinal flora and protected the intestinal barrier. Notably, the maintainer of macrophage polarization, Mincle, was activated in kidney and intestine of CKD mouse as well as in urotoxin stimulated macrophage, that was effectively inhibited by the treatment of A&P and Bifidobacterium combination. Overexpression of Mincle by genetic modification can abolish the inhibitory effects of A&P combined with Bifidobacterium on inflammation in urotoxin stimulated RAW264.7 cells. In summary, these findings demonstrated that A&P combined with Bifidobacterium can protect kidney against CKD by down-regulating macrophage inflammatory response in kidney and intestine via suppressing Mincle signaling, which provides a new insight in the treatment of CKD with traditional medicine.
Collapse
Affiliation(s)
- Tan Rui-Zhi
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Diao Hui
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Li Jian-Chun
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Zhong Xia
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Wang Xiao-Jia
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Wen Dan
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Jun-Ming
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China.,Department of Nephrology, Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wang Li
- Research Center for Integrated Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
15
|
Addai FP, Lin F, Wang T, Kosiba AA, Sheng P, Yu F, Gu J, Zhou Y, Shi H. Technical integrative approaches to cheese whey valorization towards sustainable environment. Food Funct 2020; 11:8407-8423. [PMID: 32955061 DOI: 10.1039/d0fo01484b] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Whey, a byproduct of cheese production, is often treated as an industrial dairy waste. A large volume of this product is disposed of annually due to inadequate bioconversion approaches. With its high pollutant load, disposal without pretreatment has raised a lot of environmental concerns alerting the need to seek optimal methods for adequately extracting and utilizing its organic content. In recent years, several techniques for whey valorization have emerged which may serve as interventionary measures against its environmental effects after disposal. In this review, we discuss five major approaches, by which whey can be converted into eco-friendly products, to significantly cut whey wastage. The approaches to whey valorization are therefore examined under the following perspectives: whey as a raw material for the production of bioethanol and prebiotic oligosaccharides via β-galactosidase and microbe catalyzed reactions, for the production of refined lactose as an excipient for pharmaceutical purposes, and the clinical significance of whey hydrolysates and their antifungal activity in food processing.
Collapse
Affiliation(s)
- Frank Peprah Addai
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Feng Lin
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Taotao Wang
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Anthony A Kosiba
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Pengcheng Sheng
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture, Zhejiang Institute of Freshwater Fisheries, Huzhou 313001, P. R. China
| | - Feng Yu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Jie Gu
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Yang Zhou
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, No. 301 Xuefu Road, Zhenjiang 212013, P. R. China.
| |
Collapse
|
16
|
Anthraquinone-containing compound in rhubarb prevents indole production via functional changes in gut microbiota. J Nat Med 2020; 75:116-128. [PMID: 33078328 DOI: 10.1007/s11418-020-01459-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 10/06/2020] [Indexed: 01/31/2023]
Abstract
Indole is produced from dietary tryptophan by tryptophanase in intestinal bacteria, such as Escherichia coli. In the liver, indole is converted into indoxyl sulfate, a uremic toxin and risk factor for chronic kidney disease (CKD). Probiotics and prebiotics are currently used for suppressing CKD, but there are no drugs that directly suppress indole production. In this study, we developed an optimized HPLC method for analyzing indole production and evaluated the effect of diets and rhubarb on indole production via the changes of gut microbiota. In high-carbohydrate and high-fat diet-fed mice, the indole production was significantly higher than in high-fiber diet-fed mice. We further used the high-carbohydrate diet-fed mice as a model for examining the effect of rhubarb on indole production. The 20% methanol-eluted fraction of aqueous rhubarb extract significantly suppressed indole production, and the eluate constituent rhein 8-O-β-D-glucopyranoside (RG) contributed to this effect in a concentration-dependent manner. The effect of RG depended on the anthraquinone core substructure, i.e., the aglycone moiety (rhein) of RG, which appeared to inhibit the tryptophanase function in gut microbiota. Thus, in addition to earlier reports that rhubarb is an effective CKD treatment, our study demonstrated that the anthraquinone moiety in rhubarb prevents uremic toxin production via functional changes in gut microbiota, which suppresses CKD progression.
Collapse
|
17
|
Rukavina Mikusic NL, Kouyoumdzian NM, Choi MR. Gut microbiota and chronic kidney disease: evidences and mechanisms that mediate a new communication in the gastrointestinal-renal axis. Pflugers Arch 2020; 472:303-320. [PMID: 32064574 DOI: 10.1007/s00424-020-02352-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/06/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) represents a growing public health problem associated with loss of kidney function and cardiovascular disease (CVD), the main leading cause of morbidity and mortality in CKD. It is well established that CKD is associated with gut dysbiosis. Over the past few years, there has been a growing interest in studying the composition of the gut microbiota in patients with CKD as well as the mechanisms by which gut dysbiosis contributes to CKD progression, in order to identify possible therapeutic targets to improve the morbidity and survival in CKD. The purpose of this review is to explore the clinical evidence and the mechanisms involved in the gut-kidney crosstalk as well as the possible interventions to restore a normal balance of the gut microbiota in CKD. It is well known that the influence of the gut microbiota on the gut-kidney axis acts in a reciprocal way: on the one hand, CKD significantly modifies the composition and functions of the gut microbiota. On the other hand, gut microbiota is able to manipulate the processes leading to CKD onset and progression through inflammatory, endocrine, and neurologic pathways. Understanding the complex interaction between these two organs (gut microbiota and kidney) may provide novel nephroprotective interventions to prevent the progression of CKD by targeting the gut microbiota. The review is divided into three main sections: evidences from clinical studies about the existence of a gut microbiota dysbiosis in CKD; the complex mechanisms that explain the bidirectional relationship between CKD and gut dysbiosis; and reports regarding the effects of prebiotic, probiotic, and synbiotic supplementation to restore gut microbiota balance in CKD.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Nicolás Martín Kouyoumdzian
- CONICET - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Translacional (IATIMET), Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Buenos Aires, Argentina.
- CONICET - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Translacional (IATIMET), Buenos Aires, Argentina.
| |
Collapse
|
18
|
Rational use of prebiotics for gut microbiota alterations: Specific bacterial phylotypes and related mechanisms. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103838] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
19
|
Effect of probiotics on the intestinal microbiota of hemodialysis patients: a randomized trial. Eur J Nutr 2020; 59:3755-3766. [PMID: 32112136 DOI: 10.1007/s00394-020-02207-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Intestinal dysbiosis contributes to the progression of renal failure and cardiovascular diseases in patients with chronic kidney disease. Probiotics is a promising intervention to improving intestinal dysbiosis. A double-blind clinical trial to investigate the ability of probiotics to modulate gut microbiota compositions in patients receiving hemodialysis (HD) was undertaken. METHODS Fifty HD patients were enrolled and randomized, receiving either probiotics or placebo for 6 months. The responses to the interventions on gut microbiome, serum and fecal metabolome, serum albumin and endotoxin, endothelial activation markers and inflammatory markers were assessed. RESULTS Totally, 22 in the probiotics group (11 males; 14 non-diabetic) and 23 in the placebo group (13 males; 17 non-diabetic) completed the study. Compared to that in the placebo group, probiotics did not significantly alter species diversity of the fecal microbiome. Probiotics did, however, restore the community composition, with particular significance in non-diabetic HD patients (P = 0.007 by Adonis analysis). Specifically, according to the results of linear discriminate analysis effect size, probiotics raised the proportions of family Bacteroidaceae and Enterococcaceae, and reduced Ruminococcaceae, Halomonadaceae, Peptostreptococcaceae, Clostridiales Family XIII. Incertae Sedis and Erysipelotrichaceae in non-diabetic HD patients. Additionally, probiotics reduced the abundances of several uremic retention solutes in serum or feces, including indole-3-acetic acid-O-glucuronide, 3-guanidinopropionic acid, and 1-methylinosine (P < 0.05). In the probiotic arm, no significant changes were observed in other secondary outcomes. CONCLUSIONS Taken together, outcomes from this study suggest that probiotics do have benefits on improving intestinal imbalances and lowering exposure to several uremic toxins in HD patients.
Collapse
|
20
|
Bres E, Koppe L. Is there still a place for prebiotics in chronic kidney disease? Nephrol Dial Transplant 2020; 34:1812-1816. [PMID: 31280294 DOI: 10.1093/ndt/gfz124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/20/2019] [Indexed: 12/24/2022] Open
Affiliation(s)
- Emilie Bres
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Laetitia Koppe
- Department of Nephrology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France.,University of Lyon, CarMeN lab, INSA-Lyon, INSERM U1060, INRA, Université Claude Bernard Lyon 1, Villeurbanne, France
| |
Collapse
|
21
|
Snelson M, Kellow NJ, Coughlan MT. Modulation of the Gut Microbiota by Resistant Starch as a Treatment of Chronic Kidney Diseases: Evidence of Efficacy and Mechanistic Insights. Adv Nutr 2019; 10:303-320. [PMID: 30668615 PMCID: PMC6416045 DOI: 10.1093/advances/nmy068] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/17/2018] [Accepted: 08/12/2018] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) has been associated with changes in gut microbial ecology, or "dysbiosis," which may contribute to disease progression. Recent studies have focused on dietary approaches to favorably alter the composition of the gut microbial communities as a treatment method in CKD. Resistant starch (RS), a prebiotic that promotes proliferation of gut bacteria such as Bifidobacteria and Lactobacilli, increases the production of metabolites including short-chain fatty acids, which confer a number of health-promoting benefits. However, there is a lack of mechanistic insight into how these metabolites can positively influence renal health. Emerging evidence shows that microbiota-derived metabolites can regulate the incretin axis and mitigate inflammation via expansion of regulatory T cells. Studies from animal models and patients with CKD show that RS supplementation attenuates the concentrations of uremic retention solutes, including indoxyl sulfate and p-cresyl sulfate. Here, we present the current state of knowledge linking the microbiome to CKD, we explore the efficacy of RS in animal models of CKD and in humans with the condition, and we discuss how RS supplementation could be a promising dietary approach for slowing CKD progression.
Collapse
Affiliation(s)
- Matthew Snelson
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Nicole J Kellow
- Be Active Sleep & Eat (BASE) Facility, Department of Nutrition, Dietetics, and Food, Monash University, Notting Hill, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Baker Heart Research Institute, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Lekawanvijit S. Cardiotoxicity of Uremic Toxins: A Driver of Cardiorenal Syndrome. Toxins (Basel) 2018; 10:toxins10090352. [PMID: 30200452 PMCID: PMC6162485 DOI: 10.3390/toxins10090352] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 08/19/2018] [Accepted: 08/30/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is highly prevalent in the setting of chronic kidney disease (CKD). Such coexistence of CVD and CKD—the so-called “cardiorenal or renocardiac syndrome”—contributes to exponentially increased risk of cardiovascular (CV) mortality. Uremic cardiomyopathy is a characteristic cardiac pathology commonly found in CKD. CKD patients are also predisposed to heart rhythm disorders especially atrial fibrillation. Traditional CV risk factors as well as known CKD-associated CV risk factors such as anemia are insufficient to explain CV complications in the CKD population. Accumulation of uremic retention solutes is a hallmark of impaired renal excretory function. Many of them have been considered inert solutes until their biological toxicity is unraveled and they become accepted as “uremic toxins”. Direct cardiotoxicity of uremic toxins has been increasingly demonstrated in recent years. This review offers a mechanistic insight into the pathological cardiac remodeling and dysfunction contributed by uremic toxins with a main focus on fibroblastic growth factor-23, an emerging toxin playing a central role in the chronic kidney disease–mineral bone disorder, and the two most investigated non-dialyzable protein-bound uremic toxins, indoxyl sulfate and p-cresyl sulfate. Potential therapeutic strategies that could address these toxins and their relevant mediated pathways since pre-dialysis stages are also discussed.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Rd, Sribhoom, Chiang Mai 50200, Thailand.
| |
Collapse
|
23
|
Iwashita Y, Ohya M, Yashiro M, Sonou T, Kawakami K, Nakashima Y, Yano T, Iwashita Y, Mima T, Negi S, Kubo K, Tomoda K, Odamaki T, Shigematsu T. Dietary Changes Involving Bifidobacterium longum and Other Nutrients Delays Chronic Kidney Disease Progression. Am J Nephrol 2018; 47:325-332. [PMID: 29779028 DOI: 10.1159/000488947] [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: 02/08/2018] [Accepted: 03/28/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Recent studies suggest that prebiotic and/or probiotic treatments ameliorate kidney function in humans and animals by improving the gut environment. However, the gut microbiota and kidney disease interactions remain to be determined. This study investigated whether synbiotics modulate the gut microbiota and ameliorate kidney function using a rat model of chronic kidney disease (CKD). As uremic toxins are associated with CKD-related mineral and bone disorder, the secondary aim was to evaluate the relationship between synbiotics and secondary hyperparathyroidism (SHPT). METHODS 5/6 nephrectomy (Nx) rats were developed as the CKD model. Sham-operated (sham) rats were used as the control. To investigate the effectiveness of prebiotics (glutamine, dietary fiber, and oligosaccharide) and probiotics (Bifidobacterium longum strain; GFOB diet), rats were randomly assigned to 4 groups: Nx group fed the GFOB diet (n = 10); Nx group fed the control (CON) diet (n = 10); sham group fed the GFOB diet (n = 5); and sham group fed the control diet (n = 5). Blood, feces, and kidney samples were collected and analyzed. RESULTS Serum creatinine (Cre) and blood urea nitrogen in the Nx GFOB group were significantly lower than those in the Nx CON group. Serum indoxyl sulfate in the Nx GFOB group was lower than that in the Nx CON group, and significantly correlated with serum Cre. Inorganic phosphorus and intact parathyroid hormone in the Nx GFOB group were significantly lower than those in the Nx CON group. CONCLUSION Improving the gut environment using synbiotics ameliorated kidney function and might be a pharmacological treatment for SHPT without any serious adverse events.
Collapse
Affiliation(s)
- Yuko Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Masaki Ohya
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Mitsuru Yashiro
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Tomohiro Sonou
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kazuki Kawakami
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yuri Nakashima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Takuro Yano
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Yu Iwashita
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Toru Mima
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Shigeo Negi
- Department of Nephrology, Wakayama Medical University, Wakayama, Japan
| | - Kaoru Kubo
- Laboratory Animal Research Center, Nara Medical University, Kashihara, Japan
| | - Koichi Tomoda
- Second Department of Internal Medicine, Nara Medical University, Kashihara, Japan
| | | | | |
Collapse
|
24
|
Shiba T, Makino I, Sasaki T, Fukuhara Y, Kawakami K, Kato I, Kobayashi T. p-Cresyl sulfate decreases peripheral B cells in mice with adenine-induced renal dysfunction. Toxicol Appl Pharmacol 2018; 342:50-59. [PMID: 29407365 DOI: 10.1016/j.taap.2018.01.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 12/29/2022]
Abstract
Infection is a major cause of mortality in chronic kidney disease (CKD) patients. Although immune dysfunction is a risk factor for infection in CKD patients, its causes are not fully elucidated. In the present study, we evaluated whether p-cresyl sulfate (pCS), an intestinal bacteria-derived uremic toxin, was involved in immune dysfunction in CKD. We used osmotic pumps to establish adenine-induced renal dysfunction mice with a chronically high blood pCS concentration. Analysis of lymphocyte subsets revealed that pCS significantly reduced peripheral B cells in renal dysfunction mice. In vitro, pCS inhibited interleukin (IL)-7-induced proliferation of CD43+ B-cell progenitors and suppressed IL-7-induced phosphorylation of signal transducer and activator of transcription 5 (STAT5) in these cells. Cell cycle analysis showed that pCS significantly decreased the percentage of CD43+ B-cell progenitors in S phase and increased that in G1 phase. These results suggest that pCS suppressed IL-7-induced STAT5 signaling and inhibited B-cell progenitor proliferation, leading to reduction of peripheral B cells in adenine-induced renal dysfunction mice. Therefore, pCS decreases peripheral B cells by inhibiting proliferation of CD43+ B-cell progenitors and is a likely cause of immune dysfunction in CKD patients.
Collapse
Affiliation(s)
- Takahiro Shiba
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan.
| | - Ikuyo Makino
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan
| | - Takashi Sasaki
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan
| | - Yuji Fukuhara
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan
| | - Koji Kawakami
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan
| | - Ikuo Kato
- Yakult Central Institute, 5-11 Izumi, Kunitachi-Shi, Tokyo 186-8650, Japan
| | | |
Collapse
|
25
|
Mishima E, Fukuda S, Kanemitsu Y, Saigusa D, Mukawa C, Asaji K, Matsumoto Y, Tsukamoto H, Tachikawa T, Tsukimi T, Fukuda NN, Ho HJ, Kikuchi K, Suzuki C, Nanto F, Suzuki T, Ito S, Soga T, Tomioka Y, Abe T. Canagliflozin reduces plasma uremic toxins and alters the intestinal microbiota composition in a chronic kidney disease mouse model. Am J Physiol Renal Physiol 2017; 315:F824-F833. [PMID: 29167170 DOI: 10.1152/ajprenal.00314.2017] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Accumulation of uremic toxins, which exert deleterious effects in chronic kidney disease, is influenced by the intestinal environment; the microbiota contributes to the production of representative uremic toxins, including p-cresyl sulfate and indoxyl sulfate. Canagliflozin is a sodium-glucose cotransporter (SGLT) 2 inhibitor, and it also exerts a modest inhibitory effect on SGLT1. The inhibition of intestinal SGLT1 can influence the gastrointestinal environment. We examined the effect of canagliflozin on the accumulation of uremic toxins in chronic kidney disease using adenine-induced renal failure mice. Two-week canagliflozin (10 mg/kg po) treatment did not influence the impaired renal function; however, it significantly reduced the plasma levels of p-cresyl sulfate and indoxyl sulfate in renal failure mice (a 75% and 26% reduction, respectively, compared with the vehicle group). Additionally, canagliflozin significantly increased cecal short-chain fatty acids in the mice, suggesting the promotion of bacterial carbohydrate fermentation in the intestine. Analysis of the cecal microbiota showed that canagliflozin significantly altered microbiota composition in the renal failure mice. These results indicate that canagliflozin exerts intestinal effects that reduce the accumulation of uremic toxins including p-cresyl sulfate. Reduction of accumulated uremic toxins by canagliflozin could provide a potential therapeutic option in chronic kidney disease.
Collapse
Affiliation(s)
- Eikan Mishima
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan.,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology , Kawasaki , Japan.,Transborder Medical Research Center, University of Tsukuba , Tsukuba , Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Yoshitomi Kanemitsu
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University , Sendai , Japan
| | - Chikahisa Mukawa
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Kei Asaji
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Yotaro Matsumoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Hiroki Tsukamoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Tatsuki Tachikawa
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Tomoya Tsukimi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Noriko N Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Hsin-Jung Ho
- Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan
| | - Koichi Kikuchi
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Chitose Suzuki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Fumika Nanto
- Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan
| | - Takehiro Suzuki
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Sadayoshi Ito
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Yoshihisa Tomioka
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Takaaki Abe
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine , Sendai , Japan.,Department of Medical Science, Tohoku University Graduate School of Biomedical Engineering , Sendai , Japan.,Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine , Sendai , Japan
| |
Collapse
|
26
|
Hirakawa Y, Jao TM, Inagi R. Pathophysiology and therapeutics of premature ageing in chronic kidney disease, with a focus on glycative stress. Clin Exp Pharmacol Physiol 2017; 44 Suppl 1:70-77. [PMID: 28467603 DOI: 10.1111/1440-1681.12777] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 04/12/2017] [Accepted: 04/22/2017] [Indexed: 11/30/2022]
Abstract
Chronic kidney disease (CKD) is a major concern in public health. The pathology of CKD includes premature ageing in the kidney and vessels, which results in a high risk of cardiovascular events and end-stage renal disease. Many factors are involved in premature ageing in CKD, including hormonal imbalance, glycative stress, nitrogenous metabolites, and oxidative stress. Of these, the most important role in premature ageing in CKD is played by glycative stress, namely a massive and unfavourable glycation state, since the kidney is responsible for the clearance of advanced glycation endproducts (AGEs). In an animal model, overexpression of glyoxalase I (GLO-1), a detoxifier of AGEs, has been found to alleviate premature ageing in the kidney and vessels. Both lifestyle changes and drug therapy have shown promise in overcoming premature ageing. Promising drug therapies include a GLO-1 activator and an absorbent against glycotoxin and nitrogenous metabolites.
Collapse
Affiliation(s)
- Yosuke Hirakawa
- Division of Nephrology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tzu-Ming Jao
- Division of CKD Pathophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Reiko Inagi
- Division of CKD Pathophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Koh GY, Rowling MJ. Resistant starch as a novel dietary strategy to maintain kidney health in diabetes mellitus. Nutr Rev 2017; 75:350-360. [DOI: 10.1093/nutrit/nux006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
28
|
Dietary Metabolites and Chronic Kidney Disease. Nutrients 2017; 9:nu9040358. [PMID: 28375181 PMCID: PMC5409697 DOI: 10.3390/nu9040358] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 01/04/2023] Open
Abstract
Dietary contents and their metabolites are closely related to chronic kidney disease (CKD) progression. Advanced glycated end products (AGEs) are a type of uremic toxin produced by glycation. AGE accumulation is not only the result of elevated glucose levels or reduced renal clearance capacity, but it also promotes CKD progression. Indoxyl sulfate, another uremic toxin derived from amino acid metabolism, accumulates as CKD progresses and induces tubulointerstitial fibrosis and glomerular sclerosis. Specific types of amino acids (d-serine) or fatty acids (palmitate) are reported to be closely associated with CKD progression. Promising therapeutic targets associated with nutrition include uremic toxin absorbents and inhibitors of AGEs or the receptor for AGEs (RAGE). Probiotics and prebiotics maintain gut flora balance and also prevent CKD progression by enhancing gut barriers and reducing uremic toxin formation. Nrf2 signaling not only ameliorates oxidative stress but also reduces elevated AGE levels. Bardoxolone methyl, an Nrf2 activator and NF-κB suppressor, has been tested as a therapeutic agent, but the phase 3 clinical trial was terminated owing to the high rate of cardiovascular events. However, a phase 2 trial has been initiated in Japan, and the preliminary analysis reveals promising results without an increase in cardiovascular events.
Collapse
|
29
|
Wanchai K, Pongchaidecha A, Chatsudthipong V, Chattipakorn SC, Chattipakorn N, Lungkaphin A. Role of Gastrointestinal Microbiota on Kidney Injury and the Obese Condition. Am J Med Sci 2017; 353:59-69. [DOI: 10.1016/j.amjms.2016.11.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/13/2016] [Accepted: 11/15/2016] [Indexed: 11/26/2022]
|
30
|
Ellis RJ, Small DM, Vesey DA, Johnson DW, Francis R, Vitetta L, Gobe GC, Morais C. Indoxyl sulphate and kidney disease: Causes, consequences and interventions. Nephrology (Carlton) 2016; 21:170-7. [PMID: 26239363 DOI: 10.1111/nep.12580] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2015] [Indexed: 12/28/2022]
Abstract
In the last decade, chronic kidney disease (CKD), defined as reduced renal function (glomerular filtration rate (GFR) < 60 mL/min per 1.73 m(2) ) and/or evidence of kidney damage (typically manifested as albuminuria) for at least 3 months, has become one of the fastest-growing public health concerns worldwide. CKD is characterized by reduced clearance and increased serum accumulation of metabolic waste products (uremic retention solutes). At least 152 uremic retention solutes have been reported. This review focuses on indoxyl sulphate (IS), a protein-bound, tryptophan-derived metabolite that is generated by intestinal micro-organisms (microbiota). Animal studies have demonstrated an association between IS accumulation and increased fibrosis, and oxidative stress. This has been mirrored by in vitro studies, many of which report cytotoxic effects in kidney proximal tubular cells following IS exposure. Clinical studies have associated IS accumulation with deleterious effects, such as kidney functional decline and adverse cardiovascular events, although causality has not been conclusively established. The aims of this review are to: (i) establish factors associated with increased serum accumulation of IS; (ii) report effects of IS accumulation in clinical studies; (iii) critique the reported effects of IS in the kidney, when administered both in vivo and in vitro; and (iv) summarize both established and hypothetical therapeutic options for reducing serum IS or antagonizing its reported downstream effects in the kidney.
Collapse
Affiliation(s)
- Robert J Ellis
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David M Small
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David A Vesey
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David W Johnson
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia.,Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Ross Francis
- Department of Renal Medicine, University of Queensland at Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Luis Vitetta
- Sydney Medical School - Medical Sciences, Medlab, Sydney, New South Wales, Australia.,Medlab Clinical Ltd., Medlab, Sydney, New South Wales, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Christudas Morais
- Centre for Kidney Disease Research, Translational Research Institute, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
31
|
Koh GY, Rowling MJ, Schalinske KL, Grapentine K, Loo YT. Consumption of Dietary Resistant Starch Partially Corrected the Growth Pattern Despite Hyperglycemia and Compromised Kidney Function in Streptozotocin-Induced Diabetic Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7540-7545. [PMID: 27665944 DOI: 10.1021/acs.jafc.6b03808] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We previously demonstrated that feeding of dietary resistant starch (RS) prior to the induction of diabetes delayed the progression of diabetic nephropathy and maintained vitamin D balance in streptozotocin (STZ)-induced type 1 diabetic (T1D) rats. Here, we examined the impact of RS on kidney function and vitamin D homeostasis following STZ injection. Male Sprague-Dawley rats were administered STZ and fed a standard diet containing cornstarch or 20, 10, or 5% RS for 4 weeks. T1D rats fed 10 and 20% RS, but not 5% RS, gained more weight than cornstarch-fed rats. Yet, renal health and glucose metabolism were not improved by RS. Our data suggest that RS normalized growth patterns in T1D rats after diabetes induction in a dose-dependent manner despite having no effect on blood glucose and vitamin D balances. Future interventions should focus on the preventative strategies with RS in T1D.
Collapse
Affiliation(s)
- Gar Yee Koh
- The Interdepartmental Graduate Program in Nutritional Sciences and ‡Department of Food Science and Human Nutrition, Iowa State University , Ames, Iowa 50011, United States
| | - Matthew J Rowling
- The Interdepartmental Graduate Program in Nutritional Sciences and ‡Department of Food Science and Human Nutrition, Iowa State University , Ames, Iowa 50011, United States
| | - Kevin L Schalinske
- The Interdepartmental Graduate Program in Nutritional Sciences and ‡Department of Food Science and Human Nutrition, Iowa State University , Ames, Iowa 50011, United States
| | - Kelly Grapentine
- The Interdepartmental Graduate Program in Nutritional Sciences and ‡Department of Food Science and Human Nutrition, Iowa State University , Ames, Iowa 50011, United States
| | - Yi Ting Loo
- The Interdepartmental Graduate Program in Nutritional Sciences and ‡Department of Food Science and Human Nutrition, Iowa State University , Ames, Iowa 50011, United States
| |
Collapse
|
32
|
Lekawanvijit S, Kompa AR, Krum H. Protein-bound uremic toxins: a long overlooked culprit in cardiorenal syndrome. Am J Physiol Renal Physiol 2016; 311:F52-62. [DOI: 10.1152/ajprenal.00348.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 05/01/2016] [Indexed: 11/22/2022] Open
Abstract
Protein-bound uremic toxins (PBUTs) accumulate once renal excretory function declines and are not cleared by dialysis. There is increasing evidence that PBUTs exert toxic effects on many vital organs, including the kidney, blood vessels, and heart. It has been suggested that PBUTs are likely to be a potential missing link in cardiorenal syndrome, based on the high incidence of cardiovascular events and mortality in the dialysis population, which are dramatically reduced in successful kidney transplant recipients. These data have led the call for more effective dialysis or additional adjunctive therapy to eradicate these toxins and their adverse biological effects. Indoxyl sulfate and p-cresyl sulfate are the two most problematic PBUTs, conferring renal and cardiovascular toxicity, and are derived from dietary amino acid metabolites by colonic microbial organisms. Therefore, targeting the colon where these toxins are initially produced appears to be a potential therapeutic alternative for patients with chronic kidney disease. This strategy, if approved, is likely to be applicable to predialysis patients, thereby potentially preventing progression of chronic kidney disease to end-stage renal disease as well as preventing the development of cardiorenal syndrome.
Collapse
Affiliation(s)
- Suree Lekawanvijit
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; and
| | - Andrew R. Kompa
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Henry Krum
- Centre of Cardiovascular Research and Education in Therapeutics, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| |
Collapse
|
33
|
Bindels LB, Thissen JP. Nutrition in cancer patients with cachexia: A role for the gut microbiota? CLINICAL NUTRITION EXPERIMENTAL 2016. [DOI: 10.1016/j.yclnex.2015.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Sampaio-Maia B, Simões-Silva L, Pestana M, Araujo R, Soares-Silva IJ. The Role of the Gut Microbiome on Chronic Kidney Disease. ADVANCES IN APPLIED MICROBIOLOGY 2016; 96:65-94. [PMID: 27565581 DOI: 10.1016/bs.aambs.2016.06.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) is estimated to affect nearly 500 million people worldwide and cardiovascular (CV) disease is a major cause of death in this population. However, therapeutic interventions targeting traditional CV risks are not effective at lowering the incidence of CV events or at delaying the progression of the disease in CKD patients. In recent years, disturbances of normal gut microbiome were recognized in the pathogenesis of diverse chronic diseases. Gut dysbiosis is being unraveled in CKD and pointed as a nontraditional risk factor for CV risk and CKD progression. The most often reported changes in gut microbiome in CKD are related to the lower levels of Bifidobacteriaceae and Lactobacillaceae and to higher levels of Enterobacteriaceae. Although metagenomics brought us an amplified vision on the microbial world that inhabits the human host, it still lacks the sensitivity to characterize the microbiome up to species level, not revealing alterations that occur within specific genus. Here, we review the current state-of-the-art concerning gut dysbiosis in CKD and its role in pathophysiological mechanisms in CKD, particularly in relation with CV risk. Also, the strategies towards prevention and treatment of gut dysbiosis in CKD progression will be discussed.
Collapse
Affiliation(s)
| | | | - M Pestana
- University of Porto, Porto, Portugal; São João Hospital Center, Porto, EPE, Portugal
| | - R Araujo
- University of Porto, Porto, Portugal; Flinders University, Adelaide, SA, Australia
| | | |
Collapse
|
35
|
Affiliation(s)
- Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
36
|
Varasteh S, Braber S, Garssen J, Fink-Gremmels J. Galacto-oligosaccharides exert a protective effect against heat stress in a Caco-2 cell model. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.04.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
37
|
Abstract
The essential role of the gut microbiota for health has generated tremendous interest in modulating its composition and metabolic function. One of these strategies is prebiotics, which typically refer to selectively fermented nondigestible food ingredients or substances that specifically support the growth and/or activity of health-promoting bacteria that colonize the gastrointestinal tract. In this Perspective, we argue that advances in our understanding of diet-microbiome-host interactions challenge important aspects of the current concept of prebiotics, and especially the requirement for effects to be 'selective' or 'specific'. We propose to revise this concept in an effort to shift the focus towards ecological and functional features of the microbiota more likely to be relevant for host physiology. This revision would provide a more rational basis for the identification of prebiotic compounds, and a framework by which the therapeutic potential of modulating the gut microbiota could be more fully materialized.
Collapse
|
38
|
Vanholder R, Glorieux G. The intestine and the kidneys: a bad marriage can be hazardous. Clin Kidney J 2015; 8:168-79. [PMID: 25815173 PMCID: PMC4370304 DOI: 10.1093/ckj/sfv004] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/13/2015] [Indexed: 12/20/2022] Open
Abstract
The concept that the intestine and chronic kidney disease influence each other, emerged only recently. The problem is multifaceted and bidirectional. On one hand, the composition of the intestinal microbiota impacts uraemic retention solute production, resulting in the generation of essentially protein-bound uraemic toxins with strong biological impact such as vascular damage and progression of kidney failure. On the other hand, the uraemic status affects the composition of intestinal microbiota, the generation of uraemic retention solutes and their precursors and causes disturbances in the protective epithelial barrier of the intestine and the translocation of intestinal microbiota into the body. All these elements together contribute to the disruption of the metabolic equilibrium and homeostasis typical to uraemia. Several measures with putative impact on intestinal status have recently been tested for their influence on the generation or concentration of uraemic toxins. These include dietary measures, prebiotics, probiotics, synbiotics and intestinal sorbents. Unfortunately, the quality and the evidence base of many of these studies are debatable, especially in uraemia, and often results within one study or among studies are contradictory. Nevertheless, intestinal uraemic metabolite generation remains an interesting target to obtain in the future as an alternative or additive to dialysis to decrease uraemic toxin generation. In the present review, we aim to summarize (i) the role of the intestine in uraemia by producing uraemic toxins and by generating pathophysiologically relevant changes, (ii) the role of uraemia in modifying intestinal physiology and (iii) the therapeutic options that could help to modify these effects and the studies that have assessed the impact of these therapies.
Collapse
Affiliation(s)
- Raymond Vanholder
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| | - Griet Glorieux
- Nephrology Section, 0K12 , University Hospital , Ghent B9000 , Belgium
| |
Collapse
|
39
|
Lekawanvijit S. Role of Gut-Derived Protein-Bound Uremic Toxins in Cardiorenal Syndrome and Potential Treatment Modalities. Circ J 2015; 79:2088-97. [DOI: 10.1253/circj.cj-15-0749] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|