Crimson Publishers Publish With Us Reprints e-Books Video articles

Full Text

Gastroenterology Medicine & Research

Restoring Eubiosis in Intestinal Microbiota as a Therapeutic Strategy for Chronic Kidney Disease

María Magdalena Aguirre-García1, Yoshua Espinoza-Palacios1,2, Silverio Alonso López3 and Álvaro Zamudio Tiburcio4*

1UNAM-INC Research Unit, Research Division, School of Medicine, Ignacio Chávez National Institute of Cardiology, Mexico

2Metropolitan Autonomous University-Xochimilco, Open and Distance University of Mexico, Mexico

3Department of Urologist, Chairman of Medical Specialties, Mexico

4Department of Gastroenterology, Intestinal Microbiota Transplantation, Mexico

*Corresponding author: Álvaro Zamudio Tiburcio, Department of Gastroenterology, Intestinal Microbiota Transplantation, Medical Specialties, Pennsylvania 209, Suite # 12, Col. Naples, Benito Juárez Delegation, Mexico City, Mexico

Submission:February 16, 2022;Published: February 24, 2022

DOI: 10.31031/GMR.2022.06.000644

ISSN 2637-7632
Volume6 Issue4

Abstract

Chronic Kidney Disease (CKD) is associated with intestinal dysbiosis, especially when it is accompanied by arterial hypertension, metabolic disorder, sympathetic activation, and/or immune deregulation. One particularly common mechanism of pathogenesis triggered by dysbiosis is chronic inflammation. Recent research has highlighted the importance of the microbiota and its genes in health and disease. The aim of the current contribution is to review the possible advantages of certain innovative strategies for establishing eubiosis in CKD patients. A key advance in microbiota research took place in 2007 with the characterization of the human microbiome, finding billions of bacteria in the large intestine, which is 150cm long and has a surface area of 1.3m2. Such bacteria are mainly of the phyla Bacteroidetes and Firmicutes. According to the review of the literature, the use of probiotics, prebiotics, synbiotics, postbiotics, parabiotics, and intestinal microbiota transplantation can be beneficial for CKD patients under certain conditions. When intestinal microbiota transplantation afforded a positive outcome for a patient with a C. difficile infection, the procedure was provided with greater validity because this infection is a frequent complication in CKD patients. Upon performing any procedure for reestablishing eubiosis, the recommendations of the U.S. FDA in regard to the crisis of COVID-19 must be considered.

Keywords: Chronic kidney disease; Intestinal microbiota; Intestinal microbiota transplantation; Probiotics; Prebiotics; Synbiotics

Introduction

The human microbiome was first defined by Joshua Lederberg in 2001 as the ecological community of commensal, symbiotic and pathogenic microorganisms that exists in the human organism. The microbiome had at that time largely been ignored as a determinant of health or disease, according to Lederberg [1]. The human microbiome currently refers to the set of microorganisms together with their genetic material that are found in the body. It has been given many names since 2001, including “our second genome” [2], “the invisible organ of the body” [3], “the forgotten organ” [4], “the super-organ” and “super-organism” [5], “the last human organ” [6], “the new systemic organ” [7], and “the human organ in research” [8]. In 2007, the human microbiome was characterized by the Human Microbiome Project, after an investment of ∼170 million dollars. Its constitution, and more specifically that of the Intestinal (gut) Microbiota (IM), is an important factor in Chronic Kidney Disease (CKD). The latter is a grave public health problem that affects 350 million patients around the world, with deaths ranging from 0.5 to 1 million people annually [9]. Although the microbiota is often confused with the microbiome, with the term “intestinal microbiome” used by some, they are not the same [10]. It is necessary to standardize the use of this terminology. The IM consists of over 100 billion bacteria, as well as fungi, viruses, protozoa, archaea, and other microorganisms [11]. This set of microorganisms in the intestine contains more vital genes than our own genome, evidence of its unique functions. Intestinal microorganisms aid the immune response to invasive pathogenic infections of mucous membranes and the skin. Without the IM, individuals would die from pathogens that normally cannot proliferate. Whereas eubiosis exists in healthy individuals, dysbiosis is present with many diseases, including CKD. Since each element of the IM performs specific and essential functions, dysbiosis leads to pathogenic conditions in the human organism. One particularly common mechanism of pathogenesis triggered by dysbiosis is chronic inflammation.

The gastrointestinal tract is considered the largest immune organ [12-14]. Within this context, the IM has various protective activities. It maintains intestinal homeostasis and an intact intestinal barrier, inhibits the proliferation of opportunistic pathogens (and thus the development of the corresponding diseases), modifies the microenvironmental conditions in the intestine through changes in pH, avoids the development of neoplasms, and stimulates the synthesis of vitamins and anti-inflammatory substances that our body alone cannot produce. Moreover, the IM participates in digestive functions, including the assimilation of nutrients, the production of digestive enzymes, the digestion of complex carbohydrates, the generation of certain compounds from food, the maintenance of energy balance, the supply of substrates for enterocytes, the degradation of indigestible plant polysaccharides and oxalates, and the elaboration of short-chain fatty acids derived from microbial fermentation of indigestible foods. In addition, it contributes to neurological function and development through the gut-brain axis and promotes endocrine activity (from the carbon cycle in the soil to the fermentation of food in the intestine). The IM also provides a supportive environment for pregnancy [15-24].

Many of the mechanisms of CKD tend to engender dysbiosis of the IM. For instance, the increase in urea with renal insufficiency causes a substantially greater number of bacteria [25], while changes in appetite stemming from renal failure may affect the abundance and diversity of the IM [26]. A difference was detected in the composition of the IM between CKD patients on hemodialysis and healthy controls [27]. In patients with CKD versus healthy individuals, moreover, there is a greater level of Actinobacteria, Firmicutes, and Proteobacteria phyla [28]. The colon becomes the main route for uric acid and oxalate secretion [29]. Among the possible mechanisms capable of accounting for the difference in the composition of the microbiome between patients with uremia and healthy controls is the decrease in the capacity to manage dietary fiber [30], leading to constipation in patients on hemodialysis and peritoneal dialysis [31].

Factors Involved in the Modification of the Intestinal Microbiota

The IM can be altered by stress (e.g., disease), the intake of antibiotics and some other drugs, diet, and the composition of the microbiota itself. Diet acts on the microbiota through the activity of macronutrients and micronutrients [32]. For example, polyphenols and vitamin D derived from red wine and tea modulate potentially beneficial bacteria [33]. Dietary fibers, including inulin, galactooligosaccharides, oligofructose, and arabinoxylans, promote beneficial bacteria and kill potentially harmful species [34]. In the intestine, the amount and type of fat modulate beneficial and potentially harmful microorganisms, as well as the ratio of Firmicutes to Bacteroides [35]. Meanwhile, the quantity and type of protein in the diet has a substantial and differential effect on the IM [36]. Although multiple antibiotics, with short and long-term use, generate a reduction in bacterial diversity in the IM, antibiotics have been postulated as possible adjuvant therapies in terminal CKD to diminish uremic solutes that are not effectively removed by dialysis. However, only orally administered vancomycin has shown promising results [37-39]. On the other hand, some antineoplastic drugs provide anti-carcinogenic activity by changing the composition of the microbiota [40].

Apart from diet and medications, the composition of the IM is influenced by the genetics and immune response of the host. It may also be modified by stress and at the same time contribute considerably to stress [41].

Dysbiosis (dysbacteriosis, an imbalance in the IM) participates in the genesis of many diseases [42], such as those involving inflammatory processes. It plays a major role in the mechanisms of CKD, frequently causing fatal outcomes by triggering cardiovascular events [43]. Whether with or without hemodialysis, CKD patients present dysbiosis of the IM and inflammation, usually accompanied by an increase in the level of uremic toxins, indoxyl sulfate, and p-cresyl sulfate [44]. Likewise, the prevalence of insulin resistance is higher when diabetic and obese patients exhibit a limited diversity in the IM [45]. A distinct profile of the IM has been observed in progressive IgA nephropathy, with patients displaying an elevated level of Eubacteriaceae, Ruminococcaceae, Streptococcaeae, and Lachnospiraceae [46]. In a healthy person, the fermentation of indigestible elements of food by the IM produces Short-Chain Fatty Acids (SCFA), known to protect the kidneys [47]. Apart from affecting this function, dysbiosis can alter the absorption of the intestinal barrier, leading to a series of health problems caused by greater exposure to endotoxins [48].

The composition of the IM plays a fundamental role in arterial hypertension, CKD, and the generation of nephrolithiasis [49-51]. In the case of arterial hypertension, it does so through communication with the endocrine system, the nervous system, and the immune system by regulating host homeostasis. Patients with high systolic pressure undergo changes in the composition of the IM (e.g., the abundance of Firmicutes and Bacteroidetes) [52]. Additionally, fecal and urinary dysbiosis occur in the case of calcium oxalate nephrolithiasis, which gives rise to elevated levels of Bacteroidetes spp. and Prevotella spp. compared to healthy controls [53].

The Gut-Brain-Kidney Axis

The complex interaction between the brain, the IM, and the kidneys has attracted enormous interest in recent years [54] because two-way communication is able to influence many conditions, including CKD [55], by modifying the composition and metabolism of the IM [56]. As aforementioned, an imbalance in the IM is commonly found with arterial hypertension and CKD [49-51]. Through the autonomous regulation of the brain and the signals of the intestine and kidneys, the gut-brain-kidney axis makes connections between the organs [57]. For instance, uremic toxins originate in microbial metabolism [58]. An increase in the concentration of indoxyl sulfate and p-cresyl sulfate, produced in the colon, decreases renal function. This imbalance can lead to alterations in coagulation, damage to renal tubular cells, fibrosis and cardiac hypertrophy, endothelial dysfunction, and insulin resistance [59]. Some of these toxins are bound to proteins, making it very difficult to remove them with dialysis. Furthermore, gut bacteria convert choline and betaine to trimethylamine. A recently discovered metabolite, trimethylamine N‐oxide (TMAO), is associated with atherosclerosis [60].

Therapeutic Strategies for CKD

The composition of the IM and the metabolites it produces are closely related to the existence of a healthy or diseased condition. Thus, the maintenance of homeostasis in the IM greatly helps patients suffering from CKD and arterial hypertension [61]. Possible therapeutic strategies for modulating the IM include a change in diet (e.g., an increase in fiber), the use of probiotics, prebiotics, and symbiotics, and intestinal microbiota transplantation [62,63]. Other important factors are exercise, stress management, and the avoidance of certain drugs (especially antibiotics as well as antineoplastic and anti-inflammatory drugs) [64]. Sedentarism, smoking, and inappropriate eating habits are risk factors for cardiovascular disease, which is associated with the progression of CKD.

Diet

Dietary changes as a therapeutic strategy for CKD are aimed at improving subclinical acidosis in order to preserve muscles and bones and decrease the glomerular filtration rate. The reduction of acid by increasing sodium in the diet helps to protect the kidneys [65], and the inclusion of base-inducing fruits and vegetables attenuates kidney injury [66]. However, these measures are insufficient. Monitoring urinary acid excretion may be helpful [67]. The best diet for CKD is plant-based, consisting of proteins, sodium, phosphorus, and potassium. A limited consumption of processed meats, refined sugar, and sodium is desirable, as is an elevated content of fibers and grains [68]. The Mediterranean diet usually provides benefits because of the comorbidities found with nephropathy [69]. The DASH-type diet of the Institutes of Health of the USA is also advantageous under certain conditions [70]. In addition to diet, exercise represents an essential and often overlooked therapeutic strategy [71]. Without adequate exercise, CKD patients experience a decline in the quality of life in daily activities.

Probiotics

The International Scientific Association for Probiotics and Prebiotics (ISAPP) defines probiotics as “live microorganisms that, when administered in adequate amounts, confer a health benefit to the host” [72]. Some probiotics improve human health little by little, such as Bifidobacterium longum, prepared as an enteric capsule [73]. Probiotics possess antioxidant and anti-inflammatory properties in albino rats, probably by modulating the IM [74]. After administering Lactobacillus acidophilus, Streptococcus thermophilus and Bifidobacterium longum for 6 months, a decrease was observed in serum urea. The follow-up randomized controlled trial in 22 patients failed to reduce plasma uremic toxins [75]. It is suggested that persistent alloys due to uremia, dietary regimes, and the intestinal biochemical environment generate unfavorable conditions for the symbiotic microbiota [76]. To date, there has been a lack of quality in the intervention trials related to this new therapy [77]. Various probiotics stimulate an increase in the production of the intestinal mucus barrier, demonstrating a positive effect on the immune system [78]. Through goblet cells, probiotics can also enhance the synthesis of mucin, which has the first contact with invading bacteria [79]. Despite some good outcomes stemming from the intake of probiotics and prebiotics, the European Society for Pediatric Gastroenterology, Hepatology and Nutrition recommends the use of these products only for antibiotic diarrhea and gastroenteritis [80].

Prebiotics

The ISAPP defines prebiotics as “a substrate that is selectively utilized by host microorganisms to confer a health benefit” [81]. For instance, indigestible carbohydrates positively affect beneficial colonic bacteria [82]. Glucans and fructans appear to increase the level of Lactobacillus and Bifidobacteria [83]. Moreover, prebiotics diminish several factors, such as the serum concentration of uremic toxin and p-cresol, lipid levels, the concentration of oxidative stress indicators, systemic inflammation, and the inflammatory response in the early stages of CKD. They also improve the condition of kidney failure and prevent renal osteodystrophy [84]. Indoxyl sulfate is reduced by the oral administration of the prebiotic p-inulin or oligofructose-inulin in hemodialysis patients [85,86]. Both prebiotics and probiotics modulate the IM, promoting anaerobic bacterial metabolism and a decline in host solute production (e.g., bile salts and metabolic endotoxemia) [87]. When prebiotics are degraded by the IM, short-chain fatty acids are generated and released into the bloodstream, thus affecting distant organs. The two most important groups of prebiotics in terms of human health are galactooligosaccharides and fructooligosaccharides [88].

Synbiotics

The ISAPP currently defines synbiotics as “a mixture comprising live microorganisms and substrate(s) selectively utilized by microorganisms to confer a health benefit to the host” [89]. For example, compounds resulting from the binding of probiotics and prebiotics decrease the level of plasma p-cresol [90]. Indoxyl sulfate and p-cresyl sulfate are attributed to dysbiosis in the IM, according to a randomized study. A two-month treatment with synbiotics diminished C-reactive protein, an inflammatory marker [91]. Moreover, synbiotic therapy was found to reduce the serum concentration of uremic toxins [92]. Although some doctors suggest that probiotics, prebiotics, and synbiotics should be further developed before being administered to CKD patients, others recommend their present use, adding them to control strategies [93].

Postbiotics and parabiotics

Postbiotics are defined by the ISAPP as “the preparation of inanimate microorganisms and their components to confer a health benefit to the host” [94-96]. They have afforded a better impact on health than their parent compounds, probiotics. Postbiotics mainly consist of enzymes derived from Lactobacilli and Bifidobacteria [97]. Their development in recent years as interesting tools to modulate the microbiota has not yet taken the place of intestinal microbiota transplantation [94,95]. Postbiotics produce antioxidant, antimicrobial, and immunomodulating agents, thus positively influencing the IM [98]. These agents inhibit pathogenic organisms, improve the microbial composition and inflammatory profile, and lead to weight gain [99]. They include enzymes (e.g., NADH-peroxidase and glutathione peroxidase), organic acids (e.g., propionic acid), proteins (e.g., glutathione and cell surface proteins), polysaccharides, and lipids [100].

Probiotics are a set of bacteria that are predecessors of postbiotics. The most common probiotics come from Lactobacilli and Bifidobacteria and give rise to the postbiotics L. plantarum RG14, RG11, and TL1, which are antioxidant agents [101,102]. Probably due in part to this characteristic, inflammatory processes seem to be attenuated by the use of postbiotics, especially if they is associated with dysbiosis [103]. According to various authors, bacteria must be alive in order to be effective because their activity is fundamentally metabolic [102]. Parabiotics, on the other hand, are inactive, unviable cells capable of conferring health benefits when taken in sufficient quantities. They have provided successful therapy in some cases of CKD [104].

Intestinal microbiota transplantation

A difference in the composition of bacterial phyla between healthy individuals and CKD patients has been observed. In 2020, for example, the first report was published on a decreased level of Akkermansia muciniphila in CKD patients, which led to a reduced production of interleukin (IL)-10 (an anti-inflammatory cytokine) [105-107]. New procedures have been sought that are able to modify the IM and attenuate chronic renal inflammation [106]. Intestinal microbiota transplantation seeks to restore eubiosis after CKD or other disorders have generated dysbiosis. It can be performed through various techniques, including esophagogastroduodenojejunostomy, jejunostomy, colonoscopy, naso-jejunal tube, and enema. The procedure requires an excellent donor, perfectly studied in order to avoid the spread of disease [105]. Many doctors who deal with CKD patients still consider intestinal microbiota transplantation as a controversial treatment. Additionally, it is not exempt from complications, even if they are minor. Nevertheless, the vital role of the microbiota in the human organism is well known. In the case of a kidney transplant, for instance, the microbiota regulates immunomodulation [108]. Since no cases of intestinal microbiota transplantation or sterile fecal filtration for CKD patients have been described in the literature, their use has been limited. However, when intestinal microbiota transplantation was utilized on a patient with a C. difficile infection, the positive outcome provided this procedure with greater validity because such an infection is a frequent complication in CKD patients [109]. Another factor in favor of the application of intestinal microbiota transplantation is that many of the conditions found in CKD patients are also associated with intestinal dysbiosis, including arterial hypertension, metabolic disorder, sympathetic activation, and immune deregulation [110]. Hence, intestinal microbiota transplantation is an important option to be taken into account in cases of CKD.

Acknowledgment

MMAG is grateful to the DGAPA-PAPIIT for the grant supporting this project [# IN212422]. YEP would like to thank “Dirección General de Calidad y Educación en Salud, Secretaría de Salud, México” for supporting his participation in “Programa Nacional de Servicio Social en Investigación en Salud”.

Conflicts of Interest

The authors declare that they do not have any conflict of interests.

Ethical Approval

This report does not contain any study with human or animal subjects carried out by the authors.

Informed Consent

The authors obtained written informed consent from the patients in order to develop this article.

References

  1. Lederberg J, McCray AT (2001) Ome sweet Omics-a genealogical treasury of words. Scientist 15(7): 8.
  2. Grice EA, Segre JA (2012) The human microbiome: Our second genome. Annu Rev Genomics Hum Genet 13: 151-170.
  3. Hogan AJ (2014) The morbid anatomy of the human genome: Tracing the observational and representational approaches of postwar genetics and biomedicine the william Bynum prize essay. Med Hist 58(3): 315-336.
  4. O Hara AM, Shanahan F (2006) The gut flora as a forgotten organ. EMBO Rep 7(7): 688-693.
  5. Salvucci E (2019) The human-microbiome superorganism and its modulation to restore health. int J Food Sci Nutr 70(7): 781-795.
  6. Baquero F, Nombela C (2012) The microbiome as a human organ. Clin Microbiol Infect 18(Suppl 4): 2-4.
  7. Sinha A (2020) Gut microbiome: The new organ? hormone and metabolic research. EC Endocrinology and Metabolic Research 5(2): 01-02.
  8. Anwar H, Irfan F, Hussain G, Faisal MN (2019) Gut microbiome: A new organ system in body. In book: Eukaryotic Microbiology, Publisher Intechopen, London.
  9. Webster AC, Nagler EV, Morton RL, Masson P (2016) Chronic kidney disease. The Lancet 389(10075): 1238-1252.
  10. Kho ZY, Lal SK (2018) The human gut microbiome-a potential controller of wellness and disease. Front Microbiol 9: 1835.
  11. Guinane CM, Cotter PD (2013) Role of the gut microbiota in health and chronic gastrointestinal disease: Understanding a hidden metabolic organ. Terap Adv Gastroenterol 6(4): 295-308.
  12. Takiishi T, Morales Fenero CI, Saraiva Câmara NO (2017) Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 5(4): e1373208.
  13. Alarcón P, González M, Castro É (2016) The role of gut microbiota in the regulation of the immune response. Rev Méd Chile 144(7): 910-916.
  14. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, et al. (2015) Role of the normal gut microbiota. Worl J Gastroenterol 21(29): 8787-8803.
  15. Cavallari JF, Schertzer JA (2017) Intestinal microbiota contributes to energy balance, metabolic inflammation, and insulin resistance in obesity. J Obes Metab Syndr 26(3): 161-171.
  16. Montiel-Castro AJ, González-Cervantes RM, Bravo-Ruiseco G, Pacheco-López G (2013) The microbiota-gut-brain axis: Neurobehavioral correlates, health and sociality. Front Integr Neurosci 7: 70.
  17. Garret WS, Gordon JI, Glimcher LH (2010) Homeostasis and inflammation in the intestine. Cell 140(6): 859-870.
  18. Vivarelli S, Salemi R, Candido S, Falzone L, Santagati M, et al. (2019) Gut microbiota and cancer: From pathogenesis to therapy. Cancers (Basel) 11(1): 38.
  19. Lu J, Lu L, Yu Y, Cluette-Brown J, Martin CR, Claud EC (2018) Effects of intestinal microbiota on brain development in humanized gnotobiotic mice. Scientific Reports 8: 1-16.
  20. Cani PD, Knauf C (2016) How gut microbes talk to organs: The role of endocrine and nervous routes. Mol Metab 5(9): 743-752.
  21. Hasan N, Yang H (2019) Factors affecting the composition of the gut microbiota, and its modulation. Peer J 7: e7502.
  22. Den Besten G, Van Eunen K, Groen AK, Venema K, Reijngoud DJ, et al. (2013) The role of short-chain fatty acids in the interplay between diet, gut microbiota and host energy metabolism. J Lipid Res 54(9): 2325-2340.
  23. Rowland I, Gibson G, Heinken A, Scott K, Swann J, et al. (2018) Gut microbiota functions: Metabolism of nutrients and other food components. Eur J Nutr 57(1): 1-24.
  24. Kafeshani M (2017) The gut microbiome, diet, and chronic kidney disease. Journal of Preventive Epidemiology 2(1): e05.
  25. Gouden V, Bhatt H, Jialal I (2020) Renal function tests. Stat Pearls, Florida, USA.
  26. Hobby GP, Karaduta O, Dusio GF, Singh M, Zybailov BL (2019) Chronic kidney disease and the gut microbiome. Am J Physiol Renal Physiol 316(6): F1211-F1217.
  27. Hu X, Ouyang S, Xie Y, Gong Z, Du J (2020) Characterizing the gut microbiota in patients with chronic kidney disease. Postgrad Med 132(6): 495-505.
  28. Li FX, Wang MH, Wang JP, Li RS, Zhang YQ (2019) Alterations to the gut microbiota and their correlation with inflammatory factors in chronic kidney disease. Front Cell Infect Microbiol 9: 206.
  29. Lorenz EC, Michet CJ, Milliner DS, Lieske JC (2013) Update on oxalate crystal disease. Curr Rheumatol Rep 15(7): 340.
  30. Velasquez MT, Centron P, Barrows I, Dwivedi R, Raj DS (2018) Gut microbiota and cardiovascular uremic toxicities. Toxins (Basel) 10(7): 287.
  31. Yasuda G, Shibata K, Takizawa T, Ikeda Y, Tokita Y, et al. (2002) Prevalence of constipation in continuous ambulatory peritoneal dialysis patients and comparison with hemodialysis patients. Am J Kidney Dis 39(6): 1292-1299.
  32. Yang Q, Liang Q, Balakrishnan B, Belobrajdic DP, Feng QJ, et al. (2020) Role of dietary nutrients in the modulation of gut microbiota: A narrative review. Nutrients 12(2): 381.
  33. Singh AK, Cabral C, Kumar R, Ganguly R, Rana HK, et al. (2019) Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients 11(9): 2216.
  34. Carlson JL, Erickson JM, Lloyd BB, Slavin JL (2018) Health effects and sources of prebiotic dietary fiber. Curr Dev Nutr 2(3): nzy005.
  35. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, et al. (2019) What is the healthy gut microbiota composition? a changing ecosystem across age, environment, diet, and diseases. Microorganisms 7(1): 14.
  36. Conlon MA, Bird AR (2015) The impact of diet and lifestyle on gut microbiota and human health. Nutrients 7(1): 17-44.
  37. Langdon A, Crook N, Dantas G (2016) The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genoma Med 8(1): 39.
  38. Nazzal L, Soiefer L, Chang M, Tamizuddin F, Schatoff D, et al. (2021) Effect of vancomycin on the gut microbiome and plasma concentrations of gut-derived uremic solutes. Kidney Int Rep 6(8): 2122-2133.
  39. Kimber C, Zhang S, Johnson C, West RE 3rd, Prokopienko AJ, et al. (2020) Randomized, placebo-controlled trial of rifaximin therapy for lowering gut-derived cardiovascular toxins and inflammation in CKD. Kidney360 1(11): 1206-1216.
  40. Castillo-Rodriguez E, Fernandez-Prado R, Esteras R, Perez-Gomez MV, Gracia-Iguacel C, et al. (2018) Impact of altered intestinal microbiota on chronic kidney disease progression. Toxins (Basel) 10(7): 300.
  41. Pouncey AL, Scott AJ, Alexander JL, Marchesi J, Kinross J (2018) Gut microbiota, chemotherapy and the host: The influence of the gut microbiota on cancer treatment. Ecancermedicalscience 12: 868.
  42. Schippa S, Conte MP (2014) Dysbiotic events in gut microbiota: Impact on human health. Nutrients 6(12): 5786-5805.
  43. Bryniarski M, Hamarneh F, Yacoub R (2019) The role of chronic kidney disease-associated dysbiosis in cardiovascular disease. Exp Biol Med 244(6): 514-525.
  44. Li Y, Su X, Zhang L, Liu Y, Shi M, et al. (2019) Dysbiosis of the gut microbiome is associated with CKD5 and correlated with clinical indices of the disease: A case-controlled study. J Transl Med 17(1): 228.
  45. Aydin Ö, Nieuwdorp M, Gerdes V (2018) The gut microbiome as a target for the treatment of type 2 diabetes. Curr Diab Rep 18(8): 55.
  46. De Angelis M, Montemurno E, Piccolo M, Vannini L, Lauriero G, et al. (2014) Microbiota and metabolome associated with Immunoglobulin A Nephropathy (IgAN). PLoS One 9(6): e99006.
  47. Li LZ, Tao SB, Ma L, Fu P (2019) Roles of short-chain fatty acids in kidney diseases. Chin Med J 132(10): 1228-1232.
  48. Brandl K, Schnabl (2015) Is intestinal inflammation linking dysbiosis to gut barrier dysfunction during liver disease?. Expert Rev Gastroenterol Hepatol 9(8): 1069-1076.
  49. Oyama J, Node K (2019) Gut microbiota and hypertension. Hypertension Research 42: 741-743.
  50. Ticinesi A, Nouvenne A, Chiussi G, Castaldo G, Guerra A, et al. (2020) Calcium oxalate nephrolithiasis and gut microbiota: not just a gut-kidney axis. A nutritional perspective. Nutrients 12(2): 548.
  51. Sircana A, De Michieli F, Parente R, Framarin L (2018) Gut microbiota, hypertension and chronic kidney disease: Recent advances. Pharmacol Res 144: 390-408.
  52. Jose PA, Ray D (2015) Gut microbiota in hypertension. Curr Opin Nephrol Hypertens 24(5): 403-409.
  53. Johnson EL, Heaver SL, Walters WA, Ley RE (2017) Microbiome and metabolic disease: Revisiting the bacterial phylum Bacteroidetes. J Mol Med (Berl) 95(1): 1-8.
  54. Evenepoel P, Poesen R, Maijers B (2017) The gut-kidney axis. Pediatric Nephrology 32(11): 2005-2015.
  55. Yang T, Richards EM, Pepine CJ, Raizada K (2018) The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 14(7): 442-456.
  56. Velasquez MT, Centron P, Barrows I, Dwivedi R, Raj DS (2018) Gut microbiota and cardiovascular uremic toxicities. Toxins (Basel) 10(7): 287.
  57. Yang T, Sumners EM, Pepine CJ, Rauzada MK (2018) The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 14(7): 442-446.
  58. Koppe L, Fouque D, Soulage CO (2018) The role of gut microbiota and diet on uremic retention solutes production in the context of chronic kidney disease. Toxins (Basel) 10(4): 155.
  59. Leong SC, Sirich TL (2016) Indoxyl sulfate-review of toxicity and therapeutic strategies. Toxins (Basel) 8(12): 358.
  60. Bennett BJ, De Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, et al. (2013) Trimethylamine-N-Oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab 17(1): 49-60.
  61. Guldris SC, Parra EG, Amenós AC (2017) Gut microbiota in chronic kidney disease. Nefrologia 37(1): 9-19.
  62. Wühl E, Schaefer F (2008) Therapeutic strategies to slow chronic kidney disease progression. Pediatr Nephrol 23(5): 705-716.
  63. Ku E, Lee BJ, Wei J, Matthew R (2019) Hypertension in CKD: Core curriculum 2019. Am J Kidney Dis 74(1): 120-131.
  64. Biruete A (2018) Therapies targeting gut microbiota in chronic kidney disease: Exciting ideas, but not supported by the current evidence. American Society of Nutrition. Clinical Nutrition.
  65. Goraya N, Wesson DE (2012) Dietary management of chronic kidney disease: Protein restriction and beyond. Curr Opin Nephrol Hypertens 21(6): 635-640.
  66. Scialla JJ, Anderson C AM (2013) Dietary acid load: A novel nutritional target in chronic kidney disease?. Adv Chronic Kidney Dis 20(2): 141-149.
  67. Raphael KL (2018) Metabolic acidosis and subclinical metabolic acidosis in CKD. J Am Soc Nephrol 29(2): 376-382.
  68. Chauveau P, Aparicio M, Bellizzi V, Campbell K, Hong X, et al. (2018) Mediterranean diet as the diet of choice for patients with chronic kidney disease. Nephrol Dial Transplant 33(5): 725-735.
  69. Anderson CA, Nguyen HA, Rifkin DE (2016) Nutrition interventions in chronic kidney disease. Med Clin North Am 100(6): 1265-1283.
  70. Gallieni M, Cupisti A (2016) DASH and mediterranean diets as nutritional interventions for CKD patients. Am J Kidney Dis 68(6): 828-830.
  71. Miam RH (2016) Exercise guidelines for chronic kidney disease patients. J Ren Nutr 26(4): e23-e25.
  72. Hill C, Guarner F, Reid G, Gibson G, Merenstein D, et al. (2014) The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 17(11): 506-514.
  73. Iwashita Y, Ohya M, Yashiro M, Sonou T, Kawakami K, et al. (2018) Dietary changes involving Bifidobacterium longum and other nutrients delays chronic kidney disease progression. Am J Nephrol 47(5): 325-332.
  74. Ayyanna R, Ankaiah D, Arul V (2018) Anti-inflammatory and antioxidant properties of probiotic Bacterium Lactobacillus mucosae AN1 And Lactobacillus fermentum SNR1 in wistar albino rats. Front Microbiol 9: 3063.
  75. Stampini-Oliveira-Lopes RC, Pereira-Balbino K, de Paula-Jorge M, Queiroz-Ribeiro A, Stampini-Duarte H, et al. (2018) Modulation of intestinal microbiota, control of nitrogen products and inflammation by pre/probiotics in chronic kidney disease: A systematic Review. Nutr Hosp 35(3): 722-730.
  76. Vaziri ND (2012) CKD impairs barrier function and alters microbial flora of the intestine: A major link to inflammation and uremic toxicity. Curr Opin Nephrol Hypertens 21(6): 587-592.
  77. Brüssow H (2019) Probiotics and prebiotics in clinical tests: An update. F1000Res 8: F1000-F1157.
  78. La Fata G, Weber P, Mohajeri MH (2018) Probiotics and the gut immune system: Indirect regulation. Probotics Antimicrob Proteins 10(1): 11-21.
  79. Kim JJ, Khan WI (2013) Goblet cells and mucins: Role in Innate defense in enteric infections. Pathogens 2(1): 55-70.
  80. Szajewska H, Canani RB, Guarino A, Hojsak I (2016) Probiotics for the prevention of antibiotic-associated diarrhea in children. J Pediatr Gastroenterol Nutr 62(3): 495-506.
  81. Gibson G, Hutkins R, Sanders M, Prescott S, Reimer R, et al. (2017) Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat ev Gastroenterol Hepatol 14(8): 491-502.
  82. Mafra D, Borges N, Alvarenga L, Esgalhado M, Cardozo L, et al. (2019) Dietary components that may influence the disturbed gut microbiota in chronic kidney disease. Nutrients 11(3): 496.
  83. Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA (2019) Probiotics and prebiotics in intestinal health and disease: From biology to the Clinic. Nat Rev Gastroenterol Hepatol 16(10): 605-616.
  84. Panza F, Chiara R (2017) Short-term effects of pre/probiotics on p-cresol and indoxyl-sulphate serum concentration during the various stages of chronic kidney disease and management. Archives Renal Diseases Management 3(1): 001-005.
  85. Meijers BKI, De Loor H, Bammens B, Verbeke K, Vanrenterghem Y, et al. (2009) p-Cresyl sulfate and indoxyl sulfate in hemodialysis patients. Clin J Am Soc Nephrol 4(12): 1932-1938.
  86. Fagundes RA, Soder TF, Grokoski KC, Benetti F, Mendes RH (2018) Probiotics in the treatment of chronic kidney disease: A systematic review. J Bras Nephrol 40(3): 278-286.
  87. Barathikannan K, Chelliah R, Rubab M, Daliri EBM, Elahi F, et al. (2019) Gut microbiome modulation based on probiotic application for anti-obesity: A review on efficacy and validation. Microorganisms 7(10): 456.
  88. Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, et al. (2019) Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods 8(3): 92.
  89. Swanson K, Gibson G, Hutkins R, Reimer R, Reid G, et al. (2020) The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat Rev Gastroenterol Hepatol 17: 687-701.
  90. Guida B, Cataldi M, Memoli A, Trio R, di Mauro M, et al. (2017) Effect of a short-course treatment with synbiotics on plasma p-cresol concentration in kidney transplant recipients. Journal of the American College of Nutrition 36(7): 586-591.
  91. Rossi M, Johnson DW, Morrison M, Pascoe EM, Coombs JS, et al. (2016) Synbiotics Easing Renal Failure by Improving Gut Microbiology (SYNERGY): A Randomized Trial. CJASN 11(2): 223-231.
  92. Pei M, Wei L, Hu S, Yang B, Si J, et al. (2018) Probiotics, prebiotics and synbiotics for chronic kidney disease: Protocol for a Systematic review and meta-analysis. BMJ Open 8(7): e020863.
  93. Zheng HJ, Guo J, Wang Q, Wang L, Wang Y, et al. (2021) Probiotics, prebiotics, and synbiotics for the improvement of metabolic profiles in patients with chronic kidney disease: A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 61(4): 577-598.
  94. Salminen S, Collado M, Endo A, Hill C, Lebeer S, et al. (2021) The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat Rev Gastroenterol Hepatol 18: 649-667.
  95. Fabio R, Pinto D, Giuliani G (2020) Postbiotic evolution in dermatology citation. EC Microbiology 16: 01-04.
  96. Wegh CAM, Geerlings SY, Knol J, Roeselers G, Belzer C (2019) Postbiotics and their potential applications in early life nutrition and beyond. Int J Mol Sci 20(19): 4673.
  97. Aguilar-Toalá JE, García-Varela R, García HS, Mata-Haro V, González-Córdova AF, et al. (2018) Postbiotics: An evolving term within the functional foods field. Trends in Food Science & Technology 75: 105-114.
  98. Koleilat A (2019) Beyond probiotics the postbiotics. Gastroenterol Hepatol Open Access 10(6): 324-326.
  99. Cicenia A, Scirocco A, Carabotti M, Pallota L, Marignani M, et al. (2014) Postbiotic activities of Lactobacilli-derived factors. J Clin Gastroenterol 48 Suppl 1: S18-S22.
  100. Noce A, Marrone G, Di Daniele F, Ottaviani E, Jones GW, et al. (2019) Impact of gut microbiota composition on onset and progression of chronic non-communicable diseases. Nutrients 11(5): 1073.
  101. Izzudin WI, Humam AM, Merzza A, Loh TC, Foo HL, et al. (2020) Dietary postbiotic Lactobacillus plantarum improves serum and ruminal antioxidant activity and upregulates hepatic antioxidant enzymes and ruminal barrier function in post-weaning lambs. Antioxidants 9(3): 250.
  102. Alard J, Peucelle V, Boutillier B, Breton J (2018) Postbiotic use represents therapeutic approach to improve the chronic inflammatory. Beneficial Microbes 9(2): 317-331.
  103. Tsilingiri K, Rescigno M (2012) Postbiotics: What else? Beneficial Microbes 4(1): 69-75.
  104. Akter S, Park JH, Jung HK (2020) Potential health-promoting benefits of paraprobiotics, inactivated probiotic cells. J Microbiol Biotechnol 30(4): 477-481.
  105. Ropot AV, Karamzin AM, Sergeyev OV (2020) Cultivation of the next-generation probiotic Akkermansia muciniphila, methods of its safe delivery to the intestine, and factors contributing to its growth In Vivo. Current Microbiology 77: 1363-1372.
  106. Li FX, Wang MH, Wang JP, Li RS, Zhang YQ, et al. (2019) Alterations to the gut microbiota and their correlation with inflammatory factors in chronic kidney disease. Front Cell Infect Microbiol 9: 206.
  107. Ramai D, Zakhia K, Ofosu A, Ofori E, Reddy M (2019) Fecal microbiota transplantation: Donor relation, fresh or frozen, delivery methods, cost-effectiveness. Ann Gastroenterol 32(1): 30-38.
  108. Ahmad S, Bromberg J (2016) Current status of the microbiome in renal transplantation. Curr Opin Nephrol Hypertens 25(6): 570-576.
  109. Yang T, Richards EM, Pepine CJ, Raizada MK (2018) The gut microbiota and the brain-gut-kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol 14(7): 442-456.
  110. Vasdev S, Stuckless J, Richardson V (2011) Role of the immune system in hypertension: Modulation by dietary antioxidants. Int J Angiol 20(4): 189-212.

© 2022 Álvaro Zamudio Tiburcio. This is an open access article distributed under the terms of the Creative Commons Attribution License , which permits unrestricted use, distribution, and build upon your work non-commercially.