nutrients-logo

Journal Browser

Journal Browser

Prebiotics, Probiotics and Nutrients in Cardiovascular and Kidney Disease

A special issue of Nutrients (ISSN 2072-6643). This special issue belongs to the section "Prebiotics and Probiotics".

Deadline for manuscript submissions: closed (31 July 2023) | Viewed by 26136

Special Issue Editors

Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi’an 710061, China
Interests: microbiota; hypertension; gut–brain axis; chronic kidney disease; gut–kidney axis
Special Issues, Collections and Topics in MDPI journals
Department of Dialysis, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
Interests: microbiota; chronic kidney disease; gut–kidney–heart axis; clock gene

Special Issue Information

Dear Colleagues,

Cardiovascular disease (CVD) and chronic kidney disease (CKD) are the leading cause of death and disability worldwide. Emerging evidence suggests the important role of gut microbiota in the development and progression of CVD and CKD. The association of gut dysbiosis with CVD and CKD has been determined in both rodent models and human patients. The dysbiotic hallmark in the CVD and CKD is imbalanced gut microbiota with the depletion of short-chain fatty acid-producing bacteria and an increase in uremic toxin-producing pathobionts. Accumulation of uremic toxins deteriorates multiple organs, including the vascular system, heart, and kidney. Meanwhile, recent investigations have demonstrated the potential positive effect of different prebiotics, probiotic strains, or nutrients (e.g., potassium, omega-3 fatty acids, vitamin D, protein, methylfolate, functional foods) on the pathogenic mechanisms involved in CVD and CKD, including modulation of inflammatory and immune responses, decrement of uremic toxins, and enhancement of the intestinal barrier function, in addition to a beneficial impact on gut homeostasis and dysbiosis.

This Special Issue of Nutrients, entitled “Prebiotics, Probiotics and Nutrients in Cardiovascular and Kidney Disease”, welcomes original research or reviews on the current state of research. Submissions may include (but are not limited to) the following topics:

  • The role of prebiotics and probiotics in the development and progression of CVD and CKD.
  • The effects of specific dietary intake on gut microbiota change, and their potential beneficial effects on cardiovascular and kidney health.
  • Reviews on the impact of prebiotics, probiotics, and nutrients in cardiovascular and kidney disease.

Dr. Hongbao Li
Dr. Lei Chen
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Nutrients is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • prebiotics
  • probiotics
  • nutrients
  • microbiota
  • cardiovascular disease
  • chronic kidney disease

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

4 pages, 194 KiB  
Editorial
Prebiotics, Probiotics and Nutrients in Cardiovascular and Kidney Disease
by Zitong Lei, Menglu Xu, Ying Li, Lei Chen and Hongbao Li
Nutrients 2023, 15(19), 4284; https://doi.org/10.3390/nu15194284 - 08 Oct 2023
Cited by 1 | Viewed by 1488
Abstract
Cardiovascular disease (CVD) and chronic kidney disease (CKD) are the leading causes of mortality and health burden worldwide [...] Full article

Research

Jump to: Editorial, Review

17 pages, 4124 KiB  
Article
The Effects of Specific Gut Microbiota and Metabolites on IgA Nephropathy—Based on Mendelian Randomization and Clinical Validation
by Fang Wang, Ning Li, Siming Ni, Yu Min, Kang Wei, Hongbin Sun, Yuqi Fu, Yalan Liu and Dan Lv
Nutrients 2023, 15(10), 2407; https://doi.org/10.3390/nu15102407 - 22 May 2023
Cited by 4 | Viewed by 2706
Abstract
Background: Although recent research suggests that alterations in gut microbiota and metabolites play a critical role in the pathophysiology of immunoglobulin A nephropathy (IgAN), the causal relationship between specific intestinal flora and metabolites and the risk of IgAN remains unclear. Method: This study [...] Read more.
Background: Although recent research suggests that alterations in gut microbiota and metabolites play a critical role in the pathophysiology of immunoglobulin A nephropathy (IgAN), the causal relationship between specific intestinal flora and metabolites and the risk of IgAN remains unclear. Method: This study employed Mendelian randomization (MR) to investigate the causal association between gut microbiota and IgAN. To explore potential associations between gut microbiota and various outcomes, four MR methods were applied: inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode. If the results of the four methods are inconclusive, we prefer the IVW as the primary outcome. Additionally, MR-Egger, MR-PRESSO-Global, and Cochrane’s Q tests were used to detect heterogeneity and pleiotropy. The stability of MR findings was assessed using the leave-one-out approach, and the strength of the causal relationship between exposure and outcome was tested using Bonferroni correction. Additional clinical samples were utilized to validate the results of Mendelian randomization, and the outcomes were visualized through an ROC curve, confusion matrix, and correlation analysis. Result: This study examined a total of 15 metabolites and 211 microorganisms. Among them, eight bacteria and one metabolite were found to be associated with the risk of IgAN (p < 0.05). The Bonferroni-corrected test reveals that only Class. Actinobacteria (OR: 1.20, 95% CI: 1.07–1.36, p = 0.0029) have a significant causal relationship with IgAN. According to Cochrane’s Q test, there is no substantial heterogeneity across different single-nucleotide polymorphisms (p > 0.05). Furthermore, MR-Egger and MR-PRESSO-Global tests (p > 0.05) showed no evidence of pleiotropy. No reverse causal association was found between the risk of IgAN and microbiota or metabolites (p > 0.05). Clinical specimens demonstrated the effectiveness and accuracy of Actinobacteria in distinguishing IgAN patients from those with other glomerular diseases (AUC = 0.9, 95% CI: 0.78–1.00). Additionally, our correlation analysis revealed a potential association between Actinobacteria abundance and increased albuminuria (r = 0.85) and poorer prognosis in IgAN patients (p = 0.01). Conclusion: Through MR analysis, we established a causal link between Actinobacteria and the incidence of IgAN. Moreover, clinical validation using fecal samples indicated that Actinobacteria might be associated with the onset and poorer prognosis of IgAN. This finding could provide valuable biomarkers for early, noninvasive detection of the disease and potential therapeutic targets in IgAN. Full article
Show Figures

Figure 1

13 pages, 517 KiB  
Article
Safety and Effects of Lactobacillus paracasei TISTR 2593 Supplementation on Improving Cholesterol Metabolism and Atherosclerosis-Related Parameters in Subjects with Hypercholesterolemia: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial
by Jurairat Khongrum, Pratoomporn Yingthongchai, Kongsak Boonyapranai, Wachira Wongtanasarasin, Paitoon Aobchecy, Suriya Tateing, Aree Prachansuwan, Jaruwan Sitdhipol, Kanidta Niwasabutra, Punnathon Thaveethaptaikul, Pongsathon Phapugrangkul and Pennapa Chonpathompikunlert
Nutrients 2023, 15(3), 661; https://doi.org/10.3390/nu15030661 - 28 Jan 2023
Cited by 5 | Viewed by 3505
Abstract
Probiotics have the potential as a multi-target approach to modulate hypercholesterolemia associated with premature atherosclerosis. Various strains of Lactobacillus paracasei have been reported to affect hypercholesterolemia positively. This study aimed to investigate the effects of L. paracasei TISTR 2593 on lipid profile, cholesterol [...] Read more.
Probiotics have the potential as a multi-target approach to modulate hypercholesterolemia associated with premature atherosclerosis. Various strains of Lactobacillus paracasei have been reported to affect hypercholesterolemia positively. This study aimed to investigate the effects of L. paracasei TISTR 2593 on lipid profile, cholesterol metabolism, and atherosclerosis according to the registration of Thai Clinical Trial Registry as identification number TCTR 20220917002. A total of 50 participants with hypercholesterolemia were randomly and equally assigned to consume L. paracasei TISTR 2593 or a placebo in maltodextrin capsules daily. Biomarkers of lipid profiles, oxidative stress state, inflammatory state, and other biological indicators were examined on days 0, 45, and 90. The results showed that subjects taking the L. paracasei TISTR 2593 could significantly reduce the level of serum low-density lipoprotein-cholesterol (p < 0.05), malondialdehyde (p < 0.001), and tumor necrosis factor-α (p < 0.01). Moreover, L. paracasei TISTR 2593 increased the level of serum apolipoprotein E (p < 0.01) and adiponectin (p < 0.001) significantly. No changes in serum total cholesterol, high-density lipoprotein-cholesterol, triglyceride, total bile acids, and monocyte chemoattractant protein-1 were observed during L. paracasei TISTR 2593 supplementation. Therefore, L. paracasei TISTR 2593 could be an adjuvant probiotic supplement to ameliorate hypercholesterolemia and prevent or delay the development of atherosclerosis. Full article
Show Figures

Graphical abstract

23 pages, 3447 KiB  
Article
Protection by -Biotics against Hypertension Programmed by Maternal High Fructose Diet: Rectification of Dysregulated Expression of Short-Chain Fatty Acid Receptors in the Hypothalamic Paraventricular Nucleus of Adult Offspring
by Yung-Mei Chao, You-Lin Tain, Wei-Chia Lee, Kay L. H. Wu, Hong-Ren Yu and Julie Y. H. Chan
Nutrients 2022, 14(20), 4306; https://doi.org/10.3390/nu14204306 - 14 Oct 2022
Cited by 4 | Viewed by 1862
Abstract
The role of short-chain fatty acids (SCFAs) in the brain on the developmental programming of hypertension is poorly understood. The present study explored dysregulated tissue levels of SCFAs and expression of SCFA-sensing receptors in the hypothalamic paraventricular nucleus (PVN), a key forebrain region [...] Read more.
The role of short-chain fatty acids (SCFAs) in the brain on the developmental programming of hypertension is poorly understood. The present study explored dysregulated tissue levels of SCFAs and expression of SCFA-sensing receptors in the hypothalamic paraventricular nucleus (PVN), a key forebrain region engaged in neural regulation of blood pressure of offspring to maternal high fructose diet (HFD) exposure. We further investigated the engagement of SCFA-sensing receptors in PVN in the beneficial effects of -biotics (prebiotic, probiotic, synbiotic, and postbiotic) on programmed hypertension. Maternal HFD during gestation and lactation significantly reduced circulating butyrate, along with decreased tissue level of butyrate and increased expression of SCFA-sensing receptors, GPR41 and olfr78, and tissue oxidative stress and neuroinflammation in PVN of HFD offspring that were rectified by oral supplement with -biotics. Gene silencing of GPR41 or olfr78 mRNA in PVN also protected adult HFD offspring from programmed hypertension and alleviated the induced oxidative stress and inflammation in PVN. In addition, oral supplement with postbiotic butyrate restored tissue butyrate levels, rectified expressions of GPR41 and olfr78 in PVN, and protected against programmed hypertension in adult HFD offspring. These data suggest that alterations in tissue butyrate level, expression of GPR41 and olfr78, and activation of SCFA-sensing receptor-dependent tissue oxidative stress and neuroinflammation in PVN could be novel mechanisms that underlie hypertension programmed by maternal HFD exposure in adult offspring. Furthermore, oral -biotics supplementation may exert beneficial effects on hypertension of developmental origin by targeting dysfunctional SCFA-sensing receptors in PVN to exert antioxidant and anti-inflammatory actions in the brain. Full article
Show Figures

Figure 1

21 pages, 2480 KiB  
Article
Enzobiotics—A Novel Therapy for the Elimination of Uremic Toxins in Patients with CKD (EETOX Study): A Multicenter Double-Blind Randomized Controlled Trial
by Anita Saxena, Sanjay Srinivasa, Ilangovan Veerappan, Chakko Jacob, Amol Mahaldar, Amit Gupta and Ananthasubramaniam Rajagopal
Nutrients 2022, 14(18), 3804; https://doi.org/10.3390/nu14183804 - 15 Sep 2022
Cited by 5 | Viewed by 2425
Abstract
Design, participants, setting, and measurements: Predialysis adult participants with chronic kidney disease (CKD) and mean estimated glomerular filtration rate (eGFR) <45 mL/min per 1.73 m2) were recruited in 2019 to a multicentric double-blinded randomized controlled trial of enzobiotic therapy (synbiotics and [...] Read more.
Design, participants, setting, and measurements: Predialysis adult participants with chronic kidney disease (CKD) and mean estimated glomerular filtration rate (eGFR) <45 mL/min per 1.73 m2) were recruited in 2019 to a multicentric double-blinded randomized controlled trial of enzobiotic therapy (synbiotics and proteolytic enzymes) conducted over 12 weeks. The primary objective was to evaluate the efficacy and safety of enzobiotics in reducing the generation of p-cresol sulfate (PCS) and indoxyl sulfate (IS), stabilizing renal function, and improving quality of life (QoL), while the secondary objective was to evaluate the feasibility of the diagnostic prediction of IS and PCS from CKD parameters. Results: Of the 85 patients randomized (age 48.76 years, mean eGFR 23.24 mL/min per 1.73 m2 in the placebo group; age 54.03 years, eGFR 28.93 mL/min per 1.73 m2 in the enzobiotic group), 50 completed the study. The absolute mean value of PCS increased by 12% from 19 µg/mL (Day 0) to 21 µg/mL (Day90) for the placebo group, whereas it decreased by 31% from 23 µg/mL (Day 0) to 16 µg/mL (Day 90) for the enzobiotic group. For IS, the enzobiotic group showed a decrease (6.7%) from 11,668 to 10,888 ng/mL, whereas the placebo group showed an increase (8.8%) from 11,462 to 12,466 ng/mL (Day 90). Each patient improvement ratio for Day 90/Day 0 analysis showed that enzobiotics reduced PCS by 23% (0.77, p = 0.01). IS levels remained unchanged. In the placebo group, PCS increased by 27% (1.27, p = 0.14) and IS increased by 20% (1.20, p = 0.14). The proportion of individuals beyond the risk threshold for PCS (>20 µg/mL) was 53% for the placebo group and 32% for the enzobiotic group. The corresponding levels for IS risk (threshold >20,000 ng/mL) were 35% and 24% for the placebo and enzobiotic groups, respectively. In the placebo group, eGFR decreased by 7% (Day 90) but remained stable (1.00) in the enzobiotic group. QoL as assessed by the adversity ratio decreased significantly (p = 0.00), highlighting an improvement in the enzobiotic group compared to the placebo group. The predictive equations were as follows: PCS (Day 0 = −5.97 + 0.0453 PC + 2.987 UA − 1.310 Creat; IS (Day 0) = 756 + 1143 Creat + 436.0 Creat2. Conclusion: Enzobiotics significantly reduced the PCS and IS, as well as improved the QoL. Full article
Show Figures

Figure 1

12 pages, 1055 KiB  
Article
A Proinflammatory Diet Is Associated with Higher Risk of Peripheral Artery Disease
by Heze Fan, Juan Zhou, Yuzhi Huang, Xueying Feng, Peizhu Dang, Guoliang Li and Zuyi Yuan
Nutrients 2022, 14(17), 3490; https://doi.org/10.3390/nu14173490 - 25 Aug 2022
Cited by 5 | Viewed by 2620
Abstract
Peripheral arterial disease (PAD) has a strong relationship with inflammation. However, it is unclear whether the dietary inflammatory potential is associated with PAD. We aimed to address this knowledge gap. The dietary inflammatory index (DII) was obtained using a 24-h dietary recall interview [...] Read more.
Peripheral arterial disease (PAD) has a strong relationship with inflammation. However, it is unclear whether the dietary inflammatory potential is associated with PAD. We aimed to address this knowledge gap. The dietary inflammatory index (DII) was obtained using a 24-h dietary recall interview for each individual. Logistic regression models and restricted cubic spline were performed to assess the relationship of DII with the prevalence of PAD. In addition, Spearman correlation analysis and subgroup analysis were also undertaken. In total, 5840 individuals from the 1999–2004 National Health and Nutrition Examination Survey (NHANES) were enrolled in our study. Participants in higher DII quartile tended to have higher rates of PAD. The increase in DII scores showed a positive association with PAD after fully multivariate adjustment (OR (odds ratios) = 1.094, 95% confidence interval (CI): 1.022–1.171). The multivariable-adjusted OR and 95% CI of the highest DII index quartile compared with the lowest quartile was 1.543 (95% CI: 1.116–2.133). Subgroup analysis demonstrated that the positive association between DII and PAD was persistent across population subgroups. In conclusion, we report that a proinflammatory dietary pattern is related to a higher risk of developing PAD among US adults. Full article
Show Figures

Figure 1

13 pages, 2526 KiB  
Article
Effects of Spermidine on Gut Microbiota Modulation in Experimental Abdominal Aortic Aneurysm Mice
by Shuai Liu, Yu Liu, Jiani Zhao, Pu Yang, Wei Wang and Mingmei Liao
Nutrients 2022, 14(16), 3349; https://doi.org/10.3390/nu14163349 - 16 Aug 2022
Cited by 6 | Viewed by 2814
Abstract
Accumulating evidence in recent years has demonstrated the important role of gut microbiota in maintaining cardiovascular function. However, their functions in abdominal aortic aneurysm (AAA) are largely unexplored. In this study, we established a porcine pancreatic elastase-infused experimental AAA mouse model and explored [...] Read more.
Accumulating evidence in recent years has demonstrated the important role of gut microbiota in maintaining cardiovascular function. However, their functions in abdominal aortic aneurysm (AAA) are largely unexplored. In this study, we established a porcine pancreatic elastase-infused experimental AAA mouse model and explored gut microbiota modulation using 16S rDNA sequencing. Here, we found that a significant alteration to gut microbiota composition and function occurred in AAA. The functional change in the gut microbiome revealed dysregulated biosynthesis metabolism and transport of spermidine in AAA. Furthermore, exogenous spermidine was administrated via drinking water and attenuated the progression of experimental AAA disease, which supports our recent study that spermidine alleviates systemic inflammation and AAA. These effects were associated with remitted gut microbiota dysbiosis and metabolism in AAA progression as demonstrated by 16S rDNA gene analysis. In addition, several bacterial florae, such as Bacteroides, Parabacteroides and Prevotella, were identified to be associated with the progression of AAA. Our results uncovered altered gut microbial profiles in AAA and highlighted the potential therapeutic use of spermidine in the treatment of gut microbiota dysbiosis and AAA. Full article
Show Figures

Figure 1

Review

Jump to: Editorial, Research

15 pages, 1915 KiB  
Review
Exploring a Complex Interplay: Kidney–Gut Axis in Pediatric Chronic Kidney Disease
by Adriana Mocanu, Roxana Alexandra Bogos, Tudor Ilie Lazaruc, Laura Mihaela Trandafir, Vasile Valeriu Lupu, Ileana Ioniuc, Mirabela Alecsa, Anca Ivanov, Ancuta Lupu and Iuliana Magdalena Starcea
Nutrients 2023, 15(16), 3609; https://doi.org/10.3390/nu15163609 - 17 Aug 2023
Cited by 5 | Viewed by 1944
Abstract
The human intestinal microbiota is a highly intricate structure with a crucial role in promoting health and preventing disease. It consists of diverse microbial communities that inhabit the gut and contribute to essential functions such as food digestion, nutrient synthesis, and immune system [...] Read more.
The human intestinal microbiota is a highly intricate structure with a crucial role in promoting health and preventing disease. It consists of diverse microbial communities that inhabit the gut and contribute to essential functions such as food digestion, nutrient synthesis, and immune system development. The composition and function of the gut microbiota are influenced by a variety of factors, including diet, host genetics, and environmental features. In pediatric patients, the gut microbiota is particularly dynamic and vulnerable to disruption from endogenous and exogenous factors. Recent research has focused on understanding the interaction between the gut and kidneys. In individuals with chronic kidney disease, there is often a significant disturbance in the gut microbiota. This imbalance can be attributed to factors like increased levels of harmful toxins from the gut entering the bloodstream, inflammation, and oxidative stress. This review looks at what is known about the link between a child’s gut–kidney axis, how dysbiosis, or an imbalance in the microbiome, affects chronic kidney disease, and what treatments, both pharmaceutical and non-pharmaceutical, are available for this condition. Full article
Show Figures

Graphical abstract

15 pages, 1009 KiB  
Review
Inflammatory Response: A Crucial Way for Gut Microbes to Regulate Cardiovascular Diseases
by Wen Wang, Luo-Jiang Zhu, Yue-Qi Leng, Yu-Wan Wang, Te Shi, Wei-Zhong Wang and Jia-Cen Sun
Nutrients 2023, 15(3), 607; https://doi.org/10.3390/nu15030607 - 24 Jan 2023
Cited by 11 | Viewed by 2572
Abstract
Gut microbiota is the largest and most complex microflora in the human body, which plays a crucial role in human health and disease. Over the past 20 years, the bidirectional communication between gut microbiota and extra-intestinal organs has been extensively studied. A better [...] Read more.
Gut microbiota is the largest and most complex microflora in the human body, which plays a crucial role in human health and disease. Over the past 20 years, the bidirectional communication between gut microbiota and extra-intestinal organs has been extensively studied. A better comprehension of the alternative mechanisms for physiological and pathophysiological processes could pave the way for health. Cardiovascular disease (CVD) is one of the most common diseases that seriously threatens human health. Although previous studies have shown that cardiovascular diseases, such as heart failure, hypertension, and coronary atherosclerosis, are closely related to gut microbiota, limited understanding of the complex pathogenesis leads to poor effectiveness of clinical treatment. Dysregulation of inflammation always accounts for the damaged gastrointestinal function and deranged interaction with the cardiovascular system. This review focuses on the characteristics of gut microbiota in CVD and the significance of inflammation regulation during the whole process. In addition, strategies to prevent and treat CVD through proper regulation of gut microbiota and its metabolites are also discussed. Full article
Show Figures

Figure 1

22 pages, 2797 KiB  
Review
Resistant Starch as a Dietary Intervention to Limit the Progression of Diabetic Kidney Disease
by Anna M. Drake, Melinda T. Coughlan, Claus T. Christophersen and Matthew Snelson
Nutrients 2022, 14(21), 4547; https://doi.org/10.3390/nu14214547 - 28 Oct 2022
Cited by 7 | Viewed by 2839
Abstract
Diabetes is the leading cause of kidney disease, and as the number of individuals with diabetes increases there is a concomitant increase in the prevalence of diabetic kidney disease (DKD). Diabetes contributes to the development of DKD through a number of pathways, including [...] Read more.
Diabetes is the leading cause of kidney disease, and as the number of individuals with diabetes increases there is a concomitant increase in the prevalence of diabetic kidney disease (DKD). Diabetes contributes to the development of DKD through a number of pathways, including inflammation, oxidative stress, and the gut-kidney axis, which may be amenable to dietary therapy. Resistant starch (RS) is a dietary fibre that alters the gut microbial consortium, leading to an increase in the microbial production of short chain fatty acids. Evidence from animal and human studies indicate that short chain fatty acids are able to attenuate inflammatory and oxidative stress pathways, which may mitigate the progression of DKD. In this review, we evaluate and summarise the evidence from both preclinical models of DKD and clinical trials that have utilised RS as a dietary therapy to limit the progression of DKD. Full article
Show Figures

Figure 1

Back to TopTop