Gut microbiota in kidney disease and hypertension

Gut microbiota in kidney disease and hypertension

Accepted Manuscript Title: Gut Microbiota in Kidney Disease and Hypertension Authors: C. Antza, S. Stabouli, V. Kotsis PII: DOI: Reference: S1043-661...

NAN Sizes 0 Downloads 155 Views

Accepted Manuscript Title: Gut Microbiota in Kidney Disease and Hypertension Authors: C. Antza, S. Stabouli, V. Kotsis PII: DOI: Reference:

S1043-6618(17)31319-1 https://doi.org/10.1016/j.phrs.2018.02.028 YPHRS 3840

To appear in:

Pharmacological Research

Received date: Revised date: Accepted date:

15-10-2017 20-2-2018 21-2-2018

Please cite this article as: Antza C, Stabouli S, Kotsis V.Gut Microbiota in Kidney Disease and Hypertension.Pharmacological Research https://doi.org/10.1016/j.phrs.2018.02.028 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Gut Microbiota in Kidney Disease and Hypertension C. Antza1, S. Stabouli2, V. Kotsis1

1

Hypertension Center, 3rd Department of Internal Medicine, Papageorgiou

1st Department of Pediatrics, Aristotle University Thessaloniki. Greece

Corresponding Author: Prof. V Kotsis, MD, Ph.D

U

3rd Department of Internal Medicine, Aristotle University,

SC R

2

IP T

Hospital, Aristotle University Thessaloniki, Greece

N

ESH Hypertension-24h ambulatory blood pressure monitoring center of excellence,

A

Papageorgiou Hospital,

M

Aristotle University Thessaloniki, Greece

ED

Phone number: +306974748860

PT

E-mail: [email protected]

A

CC E

Graphical abstract

1

Abstract

The human gut microbiota is being composed of more than one hundred trillion microbial cells, including aerobic and anaerobic species as well as gram-positive and negative species. Animal based evidence suggests that the change of normal gut responsible for several clinical implications including blood pressure

IP T

microbiota is

increase and kidney function reduction. Trimethylamine-N-Oxide, short-chain fatty

SC R

acids and inflammatory factors are originated from the gut microbes and may induce changes in arteries, kidneys and blood pressure. Prebiotics and probiotics change the

U

gut microbiota and may reduce high blood pressure and ameliorate chronic kidney

N

disease suggesting a new treatment target in patients for the initial stages of

A

hypertension concomitant with other life style changes such as increased physical

Abbreviations

ED

M

exercise and weight reduction to reduce cardiovascular disease complications.

BP: blood pressure, HTN: hypertension, CKD: Chronic Kidney Disease, SNS:

PT

Sympathetic Nervous System, DM: diabetes mellitus, TMAO: Trimethylamine-N-

CC E

Oxide, SCFAs: short-chain fatty acids, Olfr78: olfactory receptor 78, eGFR: estimated glomerular filtration rate, MI: myocardial infraction, RCT: randomized

A

controlled trial

Keywords: gut microbiota, microbiome, gut dysbiosis, kidney-disease, hypertension

1. Introduction The human gut contains more than one hundred trillion microbial cells. Most of the bacterial species are anaerobes (97%). Microbiota including species, such as 2

Bacteroides,

Fusobacterium,

Eubacterium,

Bifidobacterium,

Lactobacillus,

Clostridium, Escherichia Coli, Enterococcus, Staphylococcus, and Streptococcus Spp. Microbiome is host specific, advance and modified throughout lifetime according to exogenous and endogenous factors. Most common phyla are Firmicutes, Bacteroidetes and Actinobacteria. (1, 2)

IP T

Microbes that have a creative ability of products helpful for the host are

defined as ‘probiotic’ and include Lactobacillus and Bifidobacterium spp. Potentially

SC R

pathogenic organisms including aerobic enteric bacteria such as Clostridium spp and Candida albicans are portion of the microbiota. Lack of balance in the proportion or

U

distribution of gut microbes characterized by decreased numbers of the beneficial and

N

increased numbers of the harmful bacteria, such as fungi or anaerobic bacteria is

A

possible to promote many diseases. Colonization resistance is the inhibitory ability of

M

the normal gut microbes to prevent the overgrowth of invading microbes. The symbiotic nature of the host and microbes is attributed to nutrient substance exchange.

ED

The host supply food to the microbes that provide breakdown products, such as vitamins and short-chain fatty acids. The maintenance of a healthy microbiome is

PT

believed to be the key to a healthy long life. The balance between the gut microorganisms’ colonies is important for the body homeostasis and responsible for

CC E

the proper gastrointestinal tract action, food and drug metabolism. It also contributes to reduce pathogen-invasion, to stimulate the immune system and synthesis of

A

vitamins. The observation that ex-germ-free animals demand increased caloric intake to maintain the same body weight as normal animals suggest that microbiota increase host caloric profit. This can be explained by calories availability from otherwise indigestible oligosaccharides. Another possible explanation could be the increasing nutrient uptake from the intestinal epithelium from induced pancreatic lipase activity

3

and effective dietary lipids hydrolysis and finally by the upregulation of the intestinal epithelium sodium/glucose co-transporter to increase glucose uptake. Microbiota composition may also regulate appetite behavior controlling autoantibodies targeting hormones that regulate appetite. Gut microbiota was also shown to abolish lipoprotein lipase inhibition promoting fatty acid uptake into adipocytes. (3, 4) The balance of gut

IP T

microbiota species depends on many factors such as the diet, physical activity,

genetic, and epigenetic factors such as antibiotic use. Obese leptin deficient mice had

SC R

a 50% reduction in the colonies of Bacteroidetes and a correspondent increase in

those from Firmicutes a situation known as dysbiosis. Similarly in human obese

U

individuals the Bacteroidetes-to-Firmicutes ratio is changed compared to lean.(4)

N

Dysbiosis controls the production and liberation of toxins, hormones and peptides. In

A

recent years, there is evidence that suggests a possible association for the impairment

M

of kidney function and the elevation of blood pressure (BP) due to dysbiosis. (5-7)Gutderived hormones gastrin and glucagon-like peptide-1 may upregulate sodium

ED

homeostasis that link to hypertension.(8, 9) Upregulated serotonin, dopamine, and norepinephrine production may also increase BP from sympathetic activation (10)

PT

The gut microbiota are responsible for the transformation of choline and phosphatidylcholine to produce trimethylamine, which oxidizes and synthesize

CC E

ΤΜΑΟ. (11) Products of colonic fermentation includes the SCFAs acetate, butyrate and propionate that have anti-inflammatory properties which may protect from

A

vascular and kidney damage. (12) Uremic solutes p-cresol sulfate and indoxyl sulfate are microbes metabolites cleared from the circulation by tubular secretion may also interact with the cardiovascular system. (13) The aim of this review is to determine possible mechanisms by which dysbiosis may promote hypertension (HTN) and chronic kidney disease (CKD).

4

Hormones, peptides and toxins produced from gut microbes are the possible link between gut microbes and the disease. The possible role of probiotics to prevent and treat dysbiosis induced hypertension and CKD will also be discussed.

IP T

2. Animal models for the relationship between gut microbiota and hypertension or chronic kidney disease

The Dahl salt-sensitive rats become hypertensive after 3 weeks of consuming

SC R

high salt diet and die from 6 to 10 weeks after the diet initiation. Renal lesions develope after 4 weeks consumption of the high-salt diet. These rats were not

U

hypertensive until the age of 20 weeks when they consume low salt diet. The Dahl

N

salt-resistant rats are normotensive whether fed the low or high salt diet. (14) Cecal

A

specimen of Dahl salt-sensitive or resistant rats were analyzed to sequence bacteria

M

species. Bacteria of the phylum Bacteroidetes were found in higher volume in the Dahl salt-sensitive compared with the salt-resistant rats. Both strains were retained on

ED

a high-salt diet and after administration of antibiotics for ablation of microbiota, researchers found that a single bolus transplantation of cecal content from salt-

PT

resistant rats to sensitive rats was enough to elevate BP for all their life-span and

CC E

reduce the longevity of the species. Fecal bacteria of the family Veillonellaceae found at lower levels concomitant with increased plasma acetate and heptanoate. (15) Another study noticed a significant fecal decrease in microbial enrichment and

A

increased Firmicutes/Bacteroidetes ratio in the spontaneously hypertensive rat as well as other hypertension rat models i.e. the chronic angiotensin II infusion model. High BP levels were reduced with the use of minocycline which also reduced the Firmicutes/Bacteroidetes ratio.(16) Nutrition with a diet high in fiber increases gut species that generate SCFAs such as acetate. Diet rich in fiber or insert of magnesium 5

acetate in the potable water reduced BP in the deoxycorticosterone acetate-salt mice model decreasing gut dysbiosis namely the ratio of Firmicutes to Bacteroidetes and increasing the acidify species. (17) Finally, germ-free mice were found to be protected from vascular leukocyte adhesion, aortic vessel wall infiltration of neutrophils and monocytes, kidney inflammation, cardiac fibrosis and systolic dysfunction, vascular

IP T

endothelial dysfunction and attenuation of blood pressure increase in response to AngII. (18)

SC R

Nephrectomized spontaneously hypertensive rats were reported to have

decreased numbers of Lactobacillus species compared to control animals.

U

Lactobacillus populations numbers were negatively related to urinary protein

N

excretion. Lactobacillus supplementation decreased urinary protein excretion, serum

A

urea nitrogen levels and uremic toxins i.e. indoxyl and p-cresyl sulfate in the 6-week-

M

old 5/6 nephrectomized spontaneously hypertensive rats. Renal sclerosis was restored by Lactobacillus treatment in these rats.

These findings suggest that gut

ED

Lactobacillus has a possible role against the CKD progression. (19) High-fiber diet in Male Sprague-Dawley rats with adenine-induced CKD reduced cecal pH, increased

PT

diversity and Bacteroidetes-to-Firmicutes ratio. Indoxyl sulfate and p-cresol

CC E

concentrations were diminished and as a result kidney function was improved. (20)

A

3. Dysbiosis, hypertension and chronic kidney disease

The pathophysiological mechanisms of HTN are multiple and complex.

Sympathetic

nervous

system

(SNS),

renin–angiotensin–aldosterone

system,

vasopressin system, natriuretic peptides, the immune system, the adipose tissue and adipokines interact for the development of HTN. (21) HTN, obesity and diabetes mellitus (DM) are associated with the early loss of kidney function and CKD (22, 23), 6

but the high prevalence of the disease can not only be attributed to the already known factors. Life style including increased salt, alcohol and carbohydrate consumption with reduced consumption of fruits and fiber in every day food intake affects the gut microbiota. Different pathophysiological mechanisms may contribute to the

IP T

development of HTN and CKD from gut microbiota dysbiosis such as production of

uremic toxins mainly trimethylamine-N-oxide (TMAO), reduced prophylactic SCFAs,

SC R

increased sympathetic activity, enhanced inflammation and immune response,

U

reduced nitric oxide and peptides that block the angiotensin-I converting enzyme.

N

3.1 Trimethylamine-N-Oxide

A

The gut microbiota are responsible for the transformation of choline and phosphatidylcholine to produce trimethylamine (TMA), which oxidizes and

M

synthesize ΤΜΑΟ. (11) Hepatic flavin monooxidases (FMO) catalyze the conversion

carnitine

has

ED

of TMA to TMAO. A role for gut microbes in TMAO generation from dietary been

PT

Anaeroplasmataceae,

proposed

in

rats

and

Prevotellaceae,

humans.

Deferribacteraceae, Enterobacteriaceae,

Anaerococcushydrogenalis, Clostridium asparagiforme, Clostiridium hathewayi,

CC E

Clostridium sporogenes, Escherichiafergusonii Proteuspenneri, Providencia rettgeri and Edwardsiella tarda are microbiota species producing TMA. (11, 24-26)

A

Vegans/vegetarians produce significantly less TMAO from L-carnitine through a microbiota-dependent mechanism. Plasma L-carnitine levels predict additional risk for established CVD, MI, stroke or death among subjects with elevated TMAO concentrations. Mice fed with L-carnitine for an extended period of time had changed their cecal microbial composition causing an increase in TMAO synthesis and

7

atherosclerosis. Dietary supplementation of TMAO in mice with unimpaired intestinal microbe species diminishes reverse cholesterol transport in vivo promoting atherosclerosis. Atherosclerosis susceptibility can be transmitted with gut microbial transplantation from atherosclerosis-prone mice depending on TMAO levels. (27-29) In normotensive animals BP was not affected by TMAO levels, however

IP T

TMAO may prolong the hypertensive effect of Ang II suggesting a chronic upregulation of BP when a patient has increased TMAO levels. (30) TMAO and

SC R

choline levels increase in chronic kidney disease, diabetes and metabolic syndrome and these co-founders should be considered when studies were interpreted. In a cohort

U

study, patients who underwent coronary angiography for possible CAD had no

N

differences in the TMAO plasma levels when angiographically assessed coronary

A

heart disease was present or not. TMAO plasma levels could not also predict

M

cardiovascular events for 8 years follow-up, but these results were not confirmed in other studies. (31) CKD patients with increased TMAO levels had poorer long-term

ED

survival and all-cause mortality. Long term dietary habits, which are responsible for increased TMAO levels, contribute to gradual renal fibrosis and chronic kidney

PT

disease in animal models. (32) TMAO levels were higher in hemodialysis patients compared to the control group. (33) Hemodialysis Caucasian patients with TMAO

CC E

levels over the 80th population percentile had higher risk of sudden cardiac and anycause death compared to patients with the lower quintile of TMAO.(34) Elevated

A

TMAO levels were related with the number of infracted coronary arteries after adjustment of CKD stage, lipid levels and other co-factors that may associated with CAD in patients who underwent cardiovascular surgery.(35)

3.2 Short-Chain Fatty Acids

8

SCFAs are originated from non-digestible carbohydrates that pass unmetabolized the small intestine and disintegrate from gut bacteria. The amount and type of fiber in the food change gut microbiota predominant species and consequently the type and amount of SCFAs produced. Products of colonic fermentation includes the SCFAs acetate, butyrate and propionatehave with anti-inflammatory and histone

IP T

deacetylase properties. (12) Acetate, propionate and butyrate decrease TNFa release from neutrophils, NF-kB activity, suppressed IL-6 mRNA and protein release and

SC R

inhibited immune-related gene expression. (36) Indigestible in the small intestine

oligosaccharides are fructooligosaccharides, oligofructose and xylooligosaccharides.

U

These oligosaccharides are fermented in the large bowel to SCFA, which can be

N

absorbed and used by the host. After 14 days of oligosaccharide-diet, SCFAs levels

of

anaerobes

and

reduced

total

aerobes.

(37)

After

6

week

M

amounts

A

were higher in treated rats accompanied by increased Cecal bifidobacteria and total

galactooligosaccharide diet at pigs, fecal bifidobacteria and lactobacilli were

adaptation. (38)

ED

increased and SCFAs production were higher compared with production before diet

PT

Therapy with acetate, propionate, and butyrate improved renal dysfunction in an acute kidney injury model of renal ischemia-reperfusion. Low levels of local and

CC E

systemic inflammation, oxidative cellular stress, cell activation and apoptosis was suggested as the possible protecting mechanism. In addition, SCFAs improved

A

mitochondrial biogenesis which may protect kidney epithelial cells hypoxia. (39) Olfactory receptors are transmembrane G protein-coupled receptors that related to the chemosensory process. Olfactory signaling pathway found in the kidney has a functional role in filtration rate and renin release. Olfactory receptor 78 (Olfr78) interfere with renin secretion in the renal juxtaglomerular apparatus stimulated from

9

SCFAs. SCFA receptors Olfr78 and G protein-coupled receptor 41 (Gpr41) are present in smooth muscle cells of small resistance vessels that regulate BP. Propionate an ex vivo vasodilator induced an acute hypotensive effect in wild-type mice, while treatment with orally administered antibiotics reduced the gut microbial biomass and significantly increased blood pressure in Olfr78−/− animals. (40, 41) In conclusion,

IP T

increased gut production of SCFAs may reduce BP levels via chemosensory

SC R

receptors in the kidneys and the vessels.

3.3 Immune System and Inflammation

U

Gut dysbiosis is associated with gut inflammation. The main local or

N

systematic inflammation pathways are the activation of the nuclear factor- kappa B

A

and the formulation of pro-inflammatory cytokines and coexistence of acquired

M

immunosuppression that decrease immune response. (42-44) Renin-Angiotensin System is associated with increased expression of pro-inflammatory cytokines

ED

involving both the innate and the adaptive immune response. Links between Angiotensin II, the innate immune system and the pathogenesis of hypertension have

PT

been reported. (45) Angiotensin II triggering toll like receptor 4 activation in the

CC E

kidney, vasculature and central nervous system may contribute to hypertension. Chemokine receptor expression and inappropriate inflammation is enhanced in

hypertension. T lymphocytes and monocytes/macrophages are mediators of

A

hypertensive inflammation, and these cells migrate in response to several chemokines. Chemokines CCL2 and CCL5 have long been implicated in hypertension induced tissue injury. (46) Bacterial endotoxin is a lipopolysaccharide and the major outer membrane element of the gram-negative bacteria. Endotoxin is liberated when host defense

10

mechanisms or autolysis destroy bacterial cell wall. Endotoxin breaks the intestinal barrier and burst into the circulation a situation that is defined as translocation. Gut bacteria promote the inflammation due to endotoxin. (43) Elevated levels of circulating endotoxin were common in patients with advanced CKD and significantly associated with reduced survival. (47)

IP T

Gut microbiota seems to be responsible in animals for the angiotensin IIinduced HTN, vascular malfunction and kidney inflammation. (18) Hypertensive

SC R

patients have a decreased microbial richness and diversity, decreased bacteriodetes

numbers and increased inflammatory cells especially Th17 cells that produce pro-

U

inflammatory cytokines i.e. TGF-b1, TNF-α, IL-1b and IL-6. (48) SCFAs and lactate

N

downregulate the proinflammatory answer of intestinal epithelial cells which is

M

A

protective against HTN. (49)

ED

4. New targets for the treatment of hypertension and chronic kidney disease?

PT

As gut microbiota seems to be implicated to both for CKD and HTN, the regulation of gut microbial health may be help in the treatment of these two diseases

CC E

beyond the classical approach. (50) Prebiotics are non-digestible from the upper gastrointestinal tract ingredients that reach the lower gut as substrates for gut

A

microbes fermentation. Lipid metabolism was improved in rats fed with chicory extract. (51) In treated hypertensive patients, increased daily fiber and protein consumption reduced ambulatory daytime and 24 hours systolic BP. (52) Daily fiber supplementation was able to reduce diastolic BP, independently of the body weight. (53) In addition, the consumption of lupin kernel flour, a low carbohydrate-high

11

protein and fiber flour, could decrease blood pressure. (54) Another randomized, double-blind trial showed that a fiber rich diet had a moderate BP lowering effect over a six weeks period of intervention.(55) The reducing effect of prebiotics on HTN has been attributed to their ability to reduce body weight, to attenuate insulin resistance and to increased absorption of minerals such as calcium. (56-58) The prebiotic

IP T

arabinoxylan oligosaccharides could not establish an effect on microbiota derived uremic retention solutes excretion in CKD patients. (59) When prebiotics and

SC R

probiotics were used along with low protein diet in CKD patients, kidney function declined slower compared to that of patients that had the low protein diet only. (56)

U

Probiotics are products containing an adequate dose of live microbes that have

N

been proved in target-host studies to favor a health benefit when administered in

A

specific amounts. (60) Daily consumption of fermented milk products (yoghurt)

M

including Enterococcus faecium and two strains of Streptococcus thermophiles reduced systolic BP. (61) Lactobacillus plantarum 299v decreased F2-isoprostanes,

ED

interleukin 6 and reduced monocytes adhesion in heavy smokers compared to control subjects. (62) Purple sweet potato yogurt a fermented probiotic rich in γ-aminobutyric

PT

acid content showed that hypertension-induced cardiac myocyte apoptosis was decreased in treated rats. Increased levels of phosphorylated insulin-like growth

CC E

factor-I receptor was found suggesting that the purple sweet potato yogurt may attenuate cardiomyocyte apoptosis by stimulating phosphorylated insulin-like growth

A

factor-I receptor dependent survival signaling pathways. (63) Fermented milk with both Lactobacillus casei and Streptococcus thermophilus reduced systolic BP after 8 weeks of intervention and reduced total cholesterol, triglycerides levels and increased HDL levels. (64) A meta-analysis of fourteen randomized placebo-controlled trials reported that the probiotic fermented milk reduced significantly the systolic and

12

diastolic BP by 3.1 mmHg and by 1.09 mmHg respectively compared with placebo. (65) There are also evidence that the use of probiotics helps patients with CKD to decrease creatinine or uric acid levels. (66, 67) Lactobacillus casei shirota in 16 x 109 colony-forming units dose decreased more than 10% the serum urea levels in stage 3 or 4 CKD patients. (68) Residual renal function was preserved in hemodialysis

IP T

patients treated with probiotics due to their effect to reduce serum endotoxin levels

and pro-inflammatory cytokines, and to enhance anti-inflammatory cytokine IL-10

SC R

concentrations. (69, 70)

Synbiotics are diet supplements composed of both probiotics and prebiotics.

U

(71) Despite that prebiotics and probiotics alone help BP levels, an animal study

N

shows that a synbiotic supplement of Lactobacillus plantarum HEAL19 with

A

fermented blueberry could not reduce blood pressure in hypertensive rats. (72) In

ED

levels.(Table 1 and 2) (73)

M

patients with CKD, synbiotics decreased serum p-cresyl sulfate but not indoxyl sulfate

PT

5. Gaps in evidence and conclusion

CC E

The balance of gut microbiota species has a close relationship with CKD and HTN. The regulation of sodium homeostasis, upregulated production of hormones and metabolites such as p-cresol sulfate, indoxyl sulfate, TMAO and SCFAs interact

A

with the cardiovascular system (Figure 1). Treatment of gut dysbiosis with prebiotics and probiotics could be beneficial for patients with HTN at stage I or prehypertension since the BP reduction is expected to be modest. Despite some evidences from RCTs most of these studies have included small numbers of patients. Even meta-analysis included only hundreds of patients, 13

suggesting that bigger studies are needed for better interpretation and understanding of the results. Despite that there is a new area of future research as we are not really aware of what species and the doses that are beneficial to health.

There is no

evidence in the literature about the effect of gut microbiota on children with HTN or kidney dysfunction. Research could focus on the effect of prebiotics and probiotics in

with dysbiosis that had probiotic treatment has a potential interest.

IP T

children. Prevention of the development of HTN or CKD in adulthood in children

SC R

Understanding of the pathophysiological mechanisms involved in microbiota

induced hypertension may be the key for the development of new drugs in the field.

U

More evidence about fecal microbiota transplant in animals and humans is required to

A

Formatting of funding sources

N

establish microbiota effects in hypertension.

M

This research did not receive any grant from public funding agencies, commercial, or

ED

not-profit sectors.

PT

Declaration of interest

A

CC E

Conflicts of interest: None

14

References

A

CC E

PT

ED

M

A

N

U

SC R

IP T

1. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? Trends Microbiol. 2004;12(12):562-8. 2. Mariat D, Firmesse O, Levenez F, Guimaraes V, Sokol H, Dore J, et al. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009;9:123. 3. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787-803. 4. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90(3):859-904. 5. Yan Q, Gu Y, Li X, Yang W, Jia L, Chen C, et al. Alterations of the Gut Microbiome in Hypertension. Frontiers in cellular and infection microbiology. 2017;7:381. 6. Marques FZ, Mackay CR, Kaye DM. Beyond gut feelings: how the gut microbiota regulates blood pressure. Nature reviews Cardiology. 2018;15(1):20-32. 7. Al Khodor S, Reichert B, Shatat IF. The Microbiome and Blood Pressure: Can Microbes Regulate Our Blood Pressure? Frontiers in pediatrics. 2017;5:138. 8. Afsar B, Vaziri ND, Aslan G, Tarim K, Kanbay M. Gut hormones and gut microbiota: implications for kidney function and hypertension. J Am Soc Hypertens. 2016;10(12):954-61. 9. Mishima RS, Elliott AD, Sanders P, Linz D. Gastrointestinal sodium absorption, microbiome, and hypertension. Nature reviews Cardiology. 2017;14(11):693. 10. Mittal R, Debs LH, Patel AP, Nguyen D, Patel K, O'Connor G, et al. Neurotransmitters: The Critical Modulators Regulating Gut-Brain Axis. Journal of cellular physiology. 2017;232(9):2359-72. 11. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature medicine. 2013;19(5):576-85. 12. Flint HJ, Bayer EA, Rincon MT, Lamed R, White BA. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis. Nat Rev Microbiol. 2008;6(2):121-31. 13. Vanholder R, Schepers E, Pletinck A, Nagler EV, Glorieux G. The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: a systematic review. Journal of the American Society of Nephrology : JASN. 2014;25(9):1897-907. 14. Yamazaki K, Katoh H, Yamamoto N, Kurihara K, Iobe H, Sonoda J, et al. Characterization of new inbred strains of Dahl-Iwai salt-sensitive and salt-resistant rats. Laboratory animal science. 1994;44(5):462-7. 15. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, et al. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol Genomics. 2015;47(6):187-97. 16. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65(6):1331-40. 17. Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, et al. High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation. 2017;135(10):964-77. 18. Karbach SH, Schonfelder T, Brandao I, Wilms E, Hormann N, Jackel S, et al. Gut Microbiota Promote Angiotensin II-Induced Arterial Hypertension and Vascular Dysfunction. Journal of the American Heart Association. 2016;5(9). 19. Yoshifuji A, Wakino S, Irie J, Tajima T, Hasegawa K, Kanda T, et al. Gut Lactobacillus protects against the progression of renal damage by modulating the gut environment in rats. Nephrol Dial Transplant. 2016;31(3):401-12. 15

A

CC E

PT

ED

M

A

N

U

SC R

IP T

20. Kieffer DA, Piccolo BD, Vaziri ND, Liu S, Lau WL, Khazaeli M, et al. Resistant starch alters gut microbiome and metabolomic profiles concurrent with amelioration of chronic kidney disease in rats. Am J Physiol Renal Physiol. 2016;310(9):F857-71. 21. Kotsis V, Nilsson P, Grassi G, Mancia G, Redon J, Luft F, et al. New developments in the pathogenesis of obesity-induced hypertension. Journal of hypertension. 2015;33(8):1499-508. 22. Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260-72. 23. Sabatino A, Regolisti G, Cosola C, Gesualdo L, Fiaccadori E. Intestinal Microbiota in Type 2 Diabetes and Chronic Kidney Disease. Curr Diab Rep. 2017;17(3):16. 24. Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015;6(2):e02481. 25. Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A. 2012;109(52):21307-12. 26. Zhu Y, Jameson E, Crosatti M, Schafer H, Rajakumar K, Bugg TD, et al. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc Natl Acad Sci U S A. 2014;111(11):4268-73. 27. Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem. 2015;290(9):5647-60. 28. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57-63. 29. Tuso P, Stoll SR, Li WW. A plant-based diet, atherogenesis, and coronary artery disease prevention. Perm J. 2015;19(1):62-7. 30. Ufnal M, Jazwiec R, Dadlez M, Drapala A, Sikora M, Skrzypecki J. Trimethylamine-Noxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can J Cardiol. 2014;30(12):1700-5. 31. Mueller DM, Allenspach M, Othman A, Saely CH, Muendlein A, Vonbank A, et al. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis. 2015;243(2):638-44. 32. Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116(3):448-55. 33. Hai X, Landeras V, Dobre MA, DeOreo P, Meyer TW, Hostetter TH. Mechanism of Prominent Trimethylamine Oxide (TMAO) Accumulation in Hemodialysis Patients. PLoS One. 2015;10(12):e0143731. 34. Shafi T, Powe NR, Meyer TW, Hwang S, Hai X, Melamed ML, et al. Trimethylamine NOxide and Cardiovascular Events in Hemodialysis Patients. J Am Soc Nephrol. 2017;28(1):321-31. 35. Mafune A, Iwamoto T, Tsutsumi Y, Nakashima A, Yamamoto I, Yokoyama K, et al. Associations among serum trimethylamine-N-oxide (TMAO) levels, kidney function and infarcted coronary artery number in patients undergoing cardiovascular surgery: a crosssectional study. Clin Exp Nephrol. 2016;20(5):731-9. 36. Asarat M, Apostolopoulos V, Vasiljevic T, Donkor O. Short-Chain Fatty Acids Regulate Cytokines and Th17/Treg Cells in Human Peripheral Blood Mononuclear Cells in vitro. Immunological investigations. 2016;45(3):205-22.

16

A

CC E

PT

ED

M

A

N

U

SC R

IP T

37. Campbell JM, Fahey GC, Jr., Wolf BW. Selected indigestible oligosaccharides affect large bowel mass, cecal and fecal short-chain fatty acids, pH and microflora in rats. J Nutr. 1997;127(1):130-6. 38. Smiricky-Tjardes MR, Grieshop CM, Flickinger EA, Bauer LL, Fahey GC, Jr. Dietary galactooligosaccharides affect ileal and total-tract nutrient digestibility, ileal and fecal bacterial concentrations, and ileal fermentative characteristics of growing pigs. J Anim Sci. 2003;81(10):2535-45. 39. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A, Felizardo RJ, de Almeida DC, et al. Gut Bacteria Products Prevent AKI Induced by Ischemia-Reperfusion. J Am Soc Nephrol. 2015;26(8):1877-88. 40. Pluznick JL, Zou DJ, Zhang X, Yan Q, Rodriguez-Gil DJ, Eisner C, et al. Functional expression of the olfactory signaling system in the kidney. Proc Natl Acad Sci U S A. 2009;106(6):2059-64. 41. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci U S A. 2013;110(11):4410-5. 42. Anders HJ, Andersen K, Stecher B. The intestinal microbiota, a leaky gut, and abnormal immunity in kidney disease. Kidney Int. 2013;83(6):1010-6. 43. Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted interventions. Journal of the American Society of Nephrology : JASN. 2014;25(4):657-70. 44. Ardalan M, Vahed SZ. Gut microbiota and renal transplant outcome. Biomed Pharmacother. 2017;90:229-36. 45. Biancardi VC, Bomfim GF, Reis WL, Al-Gassimi S, Nunes KP. The interplay between Angiotensin II, TLR4 and hypertension. Pharmacological research. 2017;120:88-96. 46. Rudemiller NP, Crowley SD. The role of chemokines in hypertension and consequent target organ damage. Pharmacological research. 2017;119:404-11. 47. McIntyre CW, Harrison LE, Eldehni MT, Jefferies HJ, Szeto CC, John SG, et al. Circulating endotoxemia: a novel factor in systemic inflammation and cardiovascular disease in chronic kidney disease. Clin J Am Soc Nephrol. 2011;6(1):133-41. 48. Kim S, Rodriguez V, Santisteban M, Yang T, Qi Y, Raizada M, et al. 6B.07: HYPERTENSIVE PATIENTS EXHIBIT GUT MICROBIAL DYSBIOSIS AND AN INCREASE IN TH17 CELLS. J Hypertens. 2015;33 Suppl 1:e77-8. 49. Iraporda C, Errea A, Romanin DE, Cayet D, Pereyra E, Pignataro O, et al. Lactate and short chain fatty acids produced by microbial fermentation downregulate proinflammatory responses in intestinal epithelial cells and myeloid cells. Immunobiology. 2015;220(10):11619. 50. Kang Y, Cai Y. Gut microbiota and hypertension: From pathogenesis to new therapeutic strategies. Clinics and research in hepatology and gastroenterology. 2017. 51. Kim M, Shin HK. The water-soluble extract of chicory influences serum and liver lipid concentrations, cecal short-chain fatty acid concentrations and fecal lipid excretion in rats. J Nutr. 1998;128(10):1731-6. 52. Burke V, Hodgson JM, Beilin LJ, Giangiulioi N, Rogers P, Puddey IB. Dietary protein and soluble fiber reduce ambulatory blood pressure in treated hypertensives. Hypertension. 2001;38(4):821-6. 53. Eliasson K, Ryttig KR, Hylander B, Rossner S. A dietary fibre supplement in the treatment of mild hypertension. A randomized, double-blind, placebo-controlled trial. J Hypertens. 1992;10(2):195-9. 54. Lee YP, Mori TA, Puddey IB, Sipsas S, Ackland TR, Beilin LJ, et al. Effects of lupin kernel flour-enriched bread on blood pressure: a controlled intervention study. Am J Clin Nutr. 2009;89(3):766-72.

17

A

CC E

PT

ED

M

A

N

U

SC R

IP T

55. He J, Streiffer RH, Muntner P, Krousel-Wood MA, Whelton PK. Effect of dietary fiber intake on blood pressure: a randomized, double-blind, placebo-controlled trial. J Hypertens. 2004;22(1):73-80. 56. Pavan M. Influence of prebiotic and probiotic supplementation on the progression of chronic kidney disease. Minerva Urol Nefrol. 2016;68(2):222-6. 57. Saad MF, Rewers M, Selby J, Howard G, Jinagouda S, Fahmi S, et al. Insulin resistance and hypertension: the Insulin Resistance Atherosclerosis study. Hypertension. 2004;43(6):1324-31. 58. Streppel MT, Arends LR, van 't Veer P, Grobbee DE, Geleijnse JM. Dietary fiber and blood pressure: a meta-analysis of randomized placebo-controlled trials. Arch Intern Med. 2005;165(2):150-6. 59. Poesen R, Evenepoel P, de Loor H, Delcour JA, Courtin CM, Kuypers D, et al. The Influence of Prebiotic Arabinoxylan Oligosaccharides on Microbiota Derived Uremic Retention Solutes in Patients with Chronic Kidney Disease: A Randomized Controlled Trial. PLoS One. 2016;11(4):e0153893. 60. Sanders ME. Probiotics: definition, sources, selection, and uses. Clin Infect Dis. 2008;46 Suppl 2:S58-61; discussion S144-51. 61. Agerholm-Larsen L, Raben A, Haulrik N, Hansen AS, Manders M, Astrup A. Effect of 8 week intake of probiotic milk products on risk factors for cardiovascular diseases. Eur J Clin Nutr. 2000;54(4):288-97. 62. Naruszewicz M, Johansson ML, Zapolska-Downar D, Bukowska H. Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers. Am J Clin Nutr. 2002;76(6):1249-55. 63. Lin PP, Hsieh YM, Kuo WW, Lin YM, Yeh YL, Lin CC, et al. Probiotic-fermented purple sweet potato yogurt activates compensatory IGFIR/PI3K/Akt survival pathways and attenuates cardiac apoptosis in the hearts of spontaneously hypertensive rats. Int J Mol Med. 2013;32(6):1319-28. 64. Kawase M, Hashimoto H, Hosoda M, Morita H, Hosono A. Effect of administration of fermented milk containing whey protein concentrate to rats and healthy men on serum lipids and blood pressure. J Dairy Sci. 2000;83(2):255-63. 65. Dong JY, Szeto IM, Makinen K, Gao Q, Wang J, Qin LQ, et al. Effect of probiotic fermented milk on blood pressure: a meta-analysis of randomised controlled trials. Br J Nutr. 2013;110(7):1188-94. 66. Ranganathan N, Ranganathan P, Friedman EA, Joseph A, Delano B, Goldfarb DS, et al. Pilot study of probiotic dietary supplementation for promoting healthy kidney function in patients with chronic kidney disease. Adv Ther. 2010;27(9):634-47. 67. Ranganathan N, Friedman EA, Tam P, Rao V, Ranganathan P, Dheer R. Probiotic dietary supplementation in patients with stage 3 and 4 chronic kidney disease: a 6-month pilot scale trial in Canada. Curr Med Res Opin. 2009;25(8):1919-30. 68. Miranda Alatriste PV, Urbina Arronte R, Gomez Espinosa CO, Espinosa Cuevas Mde L. Effect of probiotics on human blood urea levels in patients with chronic renal failure. Nutr Hosp. 2014;29(3):582-90. 69. Natarajan R, Pechenyak B. Randomized controlled trial of strain-specific probiotic formulation (Renadyl) in dialysis patients. 2014;2014:568571. 70. Wang IK, Wu YY, Yang YF, Ting IW, Lin CC, Yen TH, et al. The effect of probiotics on serum levels of cytokine and endotoxin in peritoneal dialysis patients: a randomised, doubleblind, placebo-controlled trial. Benef Microbes. 2015;6(4):423-30. 71. Pandey KR, Naik SR, Vakil BV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol. 2015;52(12):7577-87.

18

A

CC E

PT

ED

M

A

N

U

SC R

IP T

72. Xu J, Ahren IL, Prykhodko O, Olsson C, Ahrne S, Molin G. Intake of Blueberry Fermented by Lactobacillus plantarum Affects the Gut Microbiota of L-NAME Treated Rats. Evid Based Complement Alternat Med. 2013;2013:809128. 73. Rossi M, Johnson DW, Morrison M, Pascoe EM, Coombes JS, Forbes JM, et al. Synbiotics Easing Renal Failure by Improving Gut Microbiology (SYNERGY): A Randomized Trial. Clin J Am Soc Nephrol. 2016;11(2):223-31.

19

Figure Legend: Mechanisms of microbiota induced hypertension. The imbalance between promoting and protecting microbes is essential for hypertension development. Increase caloric availability, lipids hydrolysis and glucose uptake promote obesity which is an important risk factor for hypertension. Increased gastrin and glucagon-like peptide 1 promote salt sensitivity and volume overload

IP T

hypertension. TMAO, proinflammatory cytokines and altered innate immune system promote vessels and kidneys damage causing arterial stiffening, endothelial

A

CC E

PT

ED

M

A

N

U

SC R

dysfunction and reduction in GFR promoting hypertension.

20

I N U SC R

Table 1. Design

Intervention

Clinic or 24hABPM

SBP dif

95% CI

P

DBP dif

95% CI

P

Low Fiber

24hABPM

-5.9

-8.1, -3.7

0.001

-1.4

-3, 0.2

0.112

High Protein

Low Protein

24hABPM

-5.9

-8, -3.8

0.001

-2.6

-4.2, -1

0.006

Lupin Kernel flour

White bread

24hABPM

-2.6

-4.7, -0.6

0.01

0.8

-0.5, 2.2

0.22

Fiber Probiotic Fermented Milk Fiber

Low Fiber Placebo Placebo

Clinic Both Both

-1.8 -3.10 -1.13

-4.3, 0.8 -4.63, -1.56 -2.49, 0.23

0.17 N/A N/A

-1.2 -1.09 -1.26

-3, 0.5 -2.11, -0.06 -2.04, -0.48

0.17 N/A N/A

45

He J Dong Jia-Yi55 Streppel MT48

RCT RCT Meta-analysis Meta-analysis

ED

Lee YP44

RCT

PT

Burkev

M

High Fiber 42

Control

A

Study

A

CC E

Table1. Randomized control trials and meta-analysis for the use of nutrition supplementation with prebiotics and probiotics on 24h or clinic systolic and diastolic BP values

21

I N U SC R

Design RCT

Intervention Low Protein Diet+Prebiotic+Probiotic

Control Low Protein Diet only

Ranganathan N56

RCT

Probiotic Bacterial Formulation

Ranganathan N57

RCT

Miranda Alatriste PV58

RCT

Placebo

16X109 lactobacillus casei shirota

8X109 lactobacillus casei shirota

M

Probiotic

RCT

Renadyl(Probiotic)

Placebo

Wang IK60

RCT

Probiotic

Placebo

Rossi M63

RCT

Synbiotic

Placebo

CC E A

Placebo

ED

PT

Natarajan R59

A

Study Pavan M46

Outcome GFR Blood urea nitrogen levels Creatinine levels Uric acid Quality of life Blood urea nitrogen levels Uric acid Creatinine levels

Influence + + + + + + + +

P 0.001 <0.05 >0.05 >0.05 <0.05 0.002 0.05 >0.05

Blood urea levels

+

<0.05

WBC CRP Indoxyl glucuronide Quality of life Serum TNF-a IL-5 IL-6 Endotoxin IL-10 Serum indoxyl sulfate Serum p-cresyl sulfate

+ + + + + + + + + + -

0.057 0.071 0.058 >0.05 <0.05 <0.05 <0.05 <0.05 <0.05 >0.05 <0.05 0.03

Albuminuria

Table 2. Randomized control trials of prebiotics, probiotics and synbiotics in patients with chronic kidney disease: Positive effect on kidney function, inflammation and uremic toxins

22