Salt-Responsive Metabolite, β-Hydroxybutyrate, Attenuates Hypertension

Saroj Chakraborty, Sarah Galla, Xi Cheng, Ji-Youn Yeo, Blair Mell, Vishal Singh, BengSan Yeoh, Piu Saha, Anna V Mathew, Matam Vijay-Kumar, Bina Joe, Saroj Chakraborty, Sarah Galla, Xi Cheng, Ji-Youn Yeo, Blair Mell, Vishal Singh, BengSan Yeoh, Piu Saha, Anna V Mathew, Matam Vijay-Kumar, Bina Joe

Abstract

Dietary salt reduction and exercise are lifestyle modifications for salt-sensitive hypertensives. While exercise has prominent metabolic effects, salt has an adverse effect on metabolic syndrome, of which hypertension is a hallmark. We hypothesized that dietary salt impacts metabolism in a salt-sensitive model of hypertension. An untargeted metabolomic approach demonstrates lower circulating levels of the ketone body, beta-hydroxybutyrate (βOHB), in high salt-fed hypertensive rats. Despite the high salt intake, specific rescue of βOHB levels by nutritional supplementation of its precursor, 1,3-butanediol, attenuates hypertension and protects kidney function. This beneficial effect of βOHB was likely independent of gut-microbiotal and Th17-mediated effects of salt and instead facilitated by βOHB inhibiting the renal Nlrp3 inflammasome. The juxtaposed effects of dietary salt and exercise on salt-sensitive hypertension, which decrease and increase βOHB respectively, indicate that nutritional supplementation of a precursor of βOHB provides a similar benefit to salt-sensitive hypertension as exercise.

Keywords: Nlrp3; blood pressure; hypertension; inflammasome; inflammation; ketone body; kidney; metabolomics; salt; β-hydroxybutyrate.

Conflict of interest statement

DECLARATION OF INTERESTS

B.J. and S.C. have an interest in U.S. Patent Application serial no. 62/665,690 filed on May 2, 2018 in this paper regarding nutritional intervention for salt-sensitive hypertension for 1,3-butanediol. All other authors declare no competing interests.

Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Figure 1.. Hemodynamic and Renal Responses to…
Figure 1.. Hemodynamic and Renal Responses to Salt-Loading
(A–C) Systolic (A), diastolic (B), and mean (C) arterial pressure data obtained from BP radio telemetry studies of 50- to 52-day-old rats on low (0.3% NaCl) or on high (2% NaCl) salt containing diets. Black line: data from rats on a low salt diet (n = 8). Red line: data from rats on a high salt diet (n = 8). Data points are 4 hr moving averages. *p

Figure 2.. Detection of βOHB as a…

Figure 2.. Detection of βOHB as a Metabolite Altered by Dietary Salt

(A) PCA plots…

Figure 2.. Detection of βOHB as a Metabolite Altered by Dietary Salt
(A) PCA plots of data obtained from untargeted metabolomics analysis of plasma samples. Each sphere represents a single animal. Black spheres: low salt-fed group. Red spheres: high salt-fed group. Distinct clusters of black and red spheres indicate that the groups are dissimilar in their metabolomics profiles. (B) Volcano plot of all detected metabolites by the untargeted GC-TOF-MS. Blue and red dots represent metabolites not significantly and significantly different between low and high salt-fed rats (p 1.4). +, denotes βOHB. (C) Plasma levels of βOHB. (D) Confirmation of the decreased serum βOHB levels in fasting rats on a high salt diet. *p

Figure 3.. Nutritional Intervention with βOHB Lowered…

Figure 3.. Nutritional Intervention with βOHB Lowered Hypertension

Groups of 64- to 65-day-old S rats…

Figure 3.. Nutritional Intervention with βOHB Lowered Hypertension
Groups of 64- to 65-day-old S rats were administered with (n = 12) or without (n = 10) 20% v/v of 1,3-butanediol in drinking water for 5 weeks. (A–D) Serum levels of βOHB on week 3 post administration of 1,3-butanediol (A), systolic (B), diastolic (C), and mean (D) arterial pressure data recorded by radio telemetry after 3 weeks on 1,3-butanediol. Data points are 4 hr moving averages. (E) Fasted and fed Na+/K+ ratio in serum on week 3 post administration of 1,3-butanediol. (F and G) Microalbumin (F) and microalbumin/creatinine (G) ratio in serum on week 3 post administration of 1,3-butanediol. *p

Figure 4.. Assessment of Microbiotal Profiles and…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with Low or High Salt
(A–C) Relative abundance of Lactobacilli (A), Proteobacteria (B), and Prevotella (C) in S rats on low or high salt diets. Plotted are dots representing values from each rat along with a mean ± SEM. (D) Functional analysis of microbiotal profiles. Black bars represent functional pathways enriched in the low salt-fed group. Red bars represent functional pathways enriched in the high salt-fed group.

Figure 5.. Assessment of Microbiotal Profiles and…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with High Salt or High Salt with 1,3-Butanediol
(A–D) Relative abundance of Lactobacilli (A), Proteobacteria (B), Prevotella (C), and Akkermansia (D) in fecal samples of S rats on a high salt diet either with or without nutritional intervention with 1,3-butanediol. Plotted are dots representing values from each rat along with a mean ± SEM. (E) Bars represent functional pathways. Red color: pathways enriched in the high salt group without 1,3-butanediol. Purple color: pathways enriched in the high salt group administered with 1,3-butanediol.

Figure 6.. Assessment of Renal Nlrp3 Inflammasome…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation

(A–C) Comparisons of…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation
(A–C) Comparisons of renal quantitative real-time PCR data of Nlrp3 and Casp1 (A), IL18 (B), and IL1β (C) from 1,3-butanediol-treated rats compared to the rats without the 1,3-butanediol treatment. Pgk-1, phosphoglycerokinase-1. (D and E) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (D) and low salt versus high salt fed rats (E), for comparisons of serum IL1β. (F) Same legend as (A)–(C) for Lcn2. (G and H) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (G) and low salt versus high salt fed rats (H) for comparisons of serum Lcn2; *p ≤ 0.05; **p

Figure 7.. Remission of Kidney Fibrosis, Protein…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with βOHB
(A and B) Kidney sections of high salt-fed rats treated without (A) or with (B) 20% v/v of 1,3-butanediol were stained with Masson’s trichrome. Left: i and iv displays the images at low magnification (8×). The boxed regions in the left panel were further magnified (40×) into the right panel (ii and v). Black arrows in (ii) and (v) indicate protein casts; dark blue arrows in (iii) and (vi) denote blue staining for collagen. (C–E) Bar graphs show quantitation of renal injury (n = 4/group) (C), mRNA level of genes encoding for cellular stress and tissue fibrosis (n = 8/group) (D), and UPE (urinary protein excretion) (E). (F and G) Representative images (200× magnification) of kidney sections of high salt fed rats (F) and high salt + 1,3-butanediol fed rats (G) display immunostaining for macrophages (Cd68, brown color). Arrows indicate the infiltration of macrophages in kidney sections. (H) Quantitation of macrophage numbers. Numbers are counts of macrophages; n = 10. 10× magnification fields/group. Values are expressed as mean ± SEM (*p
All figures (7)
Similar articles
Cited by
References
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, et al. (2012). Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol 8, e1002358. - PMC - PubMed
    1. Anders HJ, Suarez-Alvarez B, Grigorescu M, Foresto-Neto O, Steiger S, Desai J, Marschner JA, Honarpisheh M, Shi C, Jordan J, et al. (2018). The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 93, 656–669. - PubMed
    1. Arima S, Uto H, Ibusuki R, Kumamoto R, Tanoue S, Mawatari S, Oda K, Numata M, Fujita H, Oketani M, et al. (2014). Hypertension exacerbates liver injury and hepatic fibrosis induced by a choline-deficient L-amino aciddefined diet in rats. Int. J. Mol. Med 33, 68–76. - PubMed
    1. Bai M, Chen Y, Zhao M, Zhang Y, He JC, Huang S, Jia Z, and Zhang A (2017). NLRP3 inflammasome activation contributes to aldosteroneinduced podocyte injury. Am. J. Physiol. Renal Physiol 312, F556–F564. - PubMed
    1. Barbato JC, Koch LG, Darvish A, Cicila GT, Metting PJ, and Britton SL (1998). Spectrum of aerobic endurance running performance in eleven inbred strains of rats. J. Appl. Physiol. (1985) 85, 530–536. - PubMed
Show all 86 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 2.. Detection of βOHB as a…
Figure 2.. Detection of βOHB as a Metabolite Altered by Dietary Salt
(A) PCA plots of data obtained from untargeted metabolomics analysis of plasma samples. Each sphere represents a single animal. Black spheres: low salt-fed group. Red spheres: high salt-fed group. Distinct clusters of black and red spheres indicate that the groups are dissimilar in their metabolomics profiles. (B) Volcano plot of all detected metabolites by the untargeted GC-TOF-MS. Blue and red dots represent metabolites not significantly and significantly different between low and high salt-fed rats (p 1.4). +, denotes βOHB. (C) Plasma levels of βOHB. (D) Confirmation of the decreased serum βOHB levels in fasting rats on a high salt diet. *p

Figure 3.. Nutritional Intervention with βOHB Lowered…

Figure 3.. Nutritional Intervention with βOHB Lowered Hypertension

Groups of 64- to 65-day-old S rats…

Figure 3.. Nutritional Intervention with βOHB Lowered Hypertension
Groups of 64- to 65-day-old S rats were administered with (n = 12) or without (n = 10) 20% v/v of 1,3-butanediol in drinking water for 5 weeks. (A–D) Serum levels of βOHB on week 3 post administration of 1,3-butanediol (A), systolic (B), diastolic (C), and mean (D) arterial pressure data recorded by radio telemetry after 3 weeks on 1,3-butanediol. Data points are 4 hr moving averages. (E) Fasted and fed Na+/K+ ratio in serum on week 3 post administration of 1,3-butanediol. (F and G) Microalbumin (F) and microalbumin/creatinine (G) ratio in serum on week 3 post administration of 1,3-butanediol. *p

Figure 4.. Assessment of Microbiotal Profiles and…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with Low or High Salt
(A–C) Relative abundance of Lactobacilli (A), Proteobacteria (B), and Prevotella (C) in S rats on low or high salt diets. Plotted are dots representing values from each rat along with a mean ± SEM. (D) Functional analysis of microbiotal profiles. Black bars represent functional pathways enriched in the low salt-fed group. Red bars represent functional pathways enriched in the high salt-fed group.

Figure 5.. Assessment of Microbiotal Profiles and…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with High Salt or High Salt with 1,3-Butanediol
(A–D) Relative abundance of Lactobacilli (A), Proteobacteria (B), Prevotella (C), and Akkermansia (D) in fecal samples of S rats on a high salt diet either with or without nutritional intervention with 1,3-butanediol. Plotted are dots representing values from each rat along with a mean ± SEM. (E) Bars represent functional pathways. Red color: pathways enriched in the high salt group without 1,3-butanediol. Purple color: pathways enriched in the high salt group administered with 1,3-butanediol.

Figure 6.. Assessment of Renal Nlrp3 Inflammasome…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation

(A–C) Comparisons of…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation
(A–C) Comparisons of renal quantitative real-time PCR data of Nlrp3 and Casp1 (A), IL18 (B), and IL1β (C) from 1,3-butanediol-treated rats compared to the rats without the 1,3-butanediol treatment. Pgk-1, phosphoglycerokinase-1. (D and E) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (D) and low salt versus high salt fed rats (E), for comparisons of serum IL1β. (F) Same legend as (A)–(C) for Lcn2. (G and H) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (G) and low salt versus high salt fed rats (H) for comparisons of serum Lcn2; *p ≤ 0.05; **p

Figure 7.. Remission of Kidney Fibrosis, Protein…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with βOHB
(A and B) Kidney sections of high salt-fed rats treated without (A) or with (B) 20% v/v of 1,3-butanediol were stained with Masson’s trichrome. Left: i and iv displays the images at low magnification (8×). The boxed regions in the left panel were further magnified (40×) into the right panel (ii and v). Black arrows in (ii) and (v) indicate protein casts; dark blue arrows in (iii) and (vi) denote blue staining for collagen. (C–E) Bar graphs show quantitation of renal injury (n = 4/group) (C), mRNA level of genes encoding for cellular stress and tissue fibrosis (n = 8/group) (D), and UPE (urinary protein excretion) (E). (F and G) Representative images (200× magnification) of kidney sections of high salt fed rats (F) and high salt + 1,3-butanediol fed rats (G) display immunostaining for macrophages (Cd68, brown color). Arrows indicate the infiltration of macrophages in kidney sections. (H) Quantitation of macrophage numbers. Numbers are counts of macrophages; n = 10. 10× magnification fields/group. Values are expressed as mean ± SEM (*p
All figures (7)
Similar articles
Cited by
References
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, et al. (2012). Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol 8, e1002358. - PMC - PubMed
    1. Anders HJ, Suarez-Alvarez B, Grigorescu M, Foresto-Neto O, Steiger S, Desai J, Marschner JA, Honarpisheh M, Shi C, Jordan J, et al. (2018). The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 93, 656–669. - PubMed
    1. Arima S, Uto H, Ibusuki R, Kumamoto R, Tanoue S, Mawatari S, Oda K, Numata M, Fujita H, Oketani M, et al. (2014). Hypertension exacerbates liver injury and hepatic fibrosis induced by a choline-deficient L-amino aciddefined diet in rats. Int. J. Mol. Med 33, 68–76. - PubMed
    1. Bai M, Chen Y, Zhao M, Zhang Y, He JC, Huang S, Jia Z, and Zhang A (2017). NLRP3 inflammasome activation contributes to aldosteroneinduced podocyte injury. Am. J. Physiol. Renal Physiol 312, F556–F564. - PubMed
    1. Barbato JC, Koch LG, Darvish A, Cicila GT, Metting PJ, and Britton SL (1998). Spectrum of aerobic endurance running performance in eleven inbred strains of rats. J. Appl. Physiol. (1985) 85, 530–536. - PubMed
Show all 86 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3.. Nutritional Intervention with βOHB Lowered…
Figure 3.. Nutritional Intervention with βOHB Lowered Hypertension
Groups of 64- to 65-day-old S rats were administered with (n = 12) or without (n = 10) 20% v/v of 1,3-butanediol in drinking water for 5 weeks. (A–D) Serum levels of βOHB on week 3 post administration of 1,3-butanediol (A), systolic (B), diastolic (C), and mean (D) arterial pressure data recorded by radio telemetry after 3 weeks on 1,3-butanediol. Data points are 4 hr moving averages. (E) Fasted and fed Na+/K+ ratio in serum on week 3 post administration of 1,3-butanediol. (F and G) Microalbumin (F) and microalbumin/creatinine (G) ratio in serum on week 3 post administration of 1,3-butanediol. *p

Figure 4.. Assessment of Microbiotal Profiles and…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with Low or High Salt
(A–C) Relative abundance of Lactobacilli (A), Proteobacteria (B), and Prevotella (C) in S rats on low or high salt diets. Plotted are dots representing values from each rat along with a mean ± SEM. (D) Functional analysis of microbiotal profiles. Black bars represent functional pathways enriched in the low salt-fed group. Red bars represent functional pathways enriched in the high salt-fed group.

Figure 5.. Assessment of Microbiotal Profiles and…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal…

Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with High Salt or High Salt with 1,3-Butanediol
(A–D) Relative abundance of Lactobacilli (A), Proteobacteria (B), Prevotella (C), and Akkermansia (D) in fecal samples of S rats on a high salt diet either with or without nutritional intervention with 1,3-butanediol. Plotted are dots representing values from each rat along with a mean ± SEM. (E) Bars represent functional pathways. Red color: pathways enriched in the high salt group without 1,3-butanediol. Purple color: pathways enriched in the high salt group administered with 1,3-butanediol.

Figure 6.. Assessment of Renal Nlrp3 Inflammasome…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation

(A–C) Comparisons of…

Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation
(A–C) Comparisons of renal quantitative real-time PCR data of Nlrp3 and Casp1 (A), IL18 (B), and IL1β (C) from 1,3-butanediol-treated rats compared to the rats without the 1,3-butanediol treatment. Pgk-1, phosphoglycerokinase-1. (D and E) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (D) and low salt versus high salt fed rats (E), for comparisons of serum IL1β. (F) Same legend as (A)–(C) for Lcn2. (G and H) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (G) and low salt versus high salt fed rats (H) for comparisons of serum Lcn2; *p ≤ 0.05; **p

Figure 7.. Remission of Kidney Fibrosis, Protein…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with βOHB
(A and B) Kidney sections of high salt-fed rats treated without (A) or with (B) 20% v/v of 1,3-butanediol were stained with Masson’s trichrome. Left: i and iv displays the images at low magnification (8×). The boxed regions in the left panel were further magnified (40×) into the right panel (ii and v). Black arrows in (ii) and (v) indicate protein casts; dark blue arrows in (iii) and (vi) denote blue staining for collagen. (C–E) Bar graphs show quantitation of renal injury (n = 4/group) (C), mRNA level of genes encoding for cellular stress and tissue fibrosis (n = 8/group) (D), and UPE (urinary protein excretion) (E). (F and G) Representative images (200× magnification) of kidney sections of high salt fed rats (F) and high salt + 1,3-butanediol fed rats (G) display immunostaining for macrophages (Cd68, brown color). Arrows indicate the infiltration of macrophages in kidney sections. (H) Quantitation of macrophage numbers. Numbers are counts of macrophages; n = 10. 10× magnification fields/group. Values are expressed as mean ± SEM (*p
All figures (7)
Similar articles
Cited by
References
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, et al. (2012). Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol 8, e1002358. - PMC - PubMed
    1. Anders HJ, Suarez-Alvarez B, Grigorescu M, Foresto-Neto O, Steiger S, Desai J, Marschner JA, Honarpisheh M, Shi C, Jordan J, et al. (2018). The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 93, 656–669. - PubMed
    1. Arima S, Uto H, Ibusuki R, Kumamoto R, Tanoue S, Mawatari S, Oda K, Numata M, Fujita H, Oketani M, et al. (2014). Hypertension exacerbates liver injury and hepatic fibrosis induced by a choline-deficient L-amino aciddefined diet in rats. Int. J. Mol. Med 33, 68–76. - PubMed
    1. Bai M, Chen Y, Zhao M, Zhang Y, He JC, Huang S, Jia Z, and Zhang A (2017). NLRP3 inflammasome activation contributes to aldosteroneinduced podocyte injury. Am. J. Physiol. Renal Physiol 312, F556–F564. - PubMed
    1. Barbato JC, Koch LG, Darvish A, Cicila GT, Metting PJ, and Britton SL (1998). Spectrum of aerobic endurance running performance in eleven inbred strains of rats. J. Appl. Physiol. (1985) 85, 530–536. - PubMed
Show all 86 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4.. Assessment of Microbiotal Profiles and…
Figure 4.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with Low or High Salt
(A–C) Relative abundance of Lactobacilli (A), Proteobacteria (B), and Prevotella (C) in S rats on low or high salt diets. Plotted are dots representing values from each rat along with a mean ± SEM. (D) Functional analysis of microbiotal profiles. Black bars represent functional pathways enriched in the low salt-fed group. Red bars represent functional pathways enriched in the high salt-fed group.
Figure 5.. Assessment of Microbiotal Profiles and…
Figure 5.. Assessment of Microbiotal Profiles and Functional Data from 16S RNA Sequencing of Fecal Samples from S Rats Fed with High Salt or High Salt with 1,3-Butanediol
(A–D) Relative abundance of Lactobacilli (A), Proteobacteria (B), Prevotella (C), and Akkermansia (D) in fecal samples of S rats on a high salt diet either with or without nutritional intervention with 1,3-butanediol. Plotted are dots representing values from each rat along with a mean ± SEM. (E) Bars represent functional pathways. Red color: pathways enriched in the high salt group without 1,3-butanediol. Purple color: pathways enriched in the high salt group administered with 1,3-butanediol.
Figure 6.. Assessment of Renal Nlrp3 Inflammasome…
Figure 6.. Assessment of Renal Nlrp3 Inflammasome and Related Measures of Inflammation
(A–C) Comparisons of renal quantitative real-time PCR data of Nlrp3 and Casp1 (A), IL18 (B), and IL1β (C) from 1,3-butanediol-treated rats compared to the rats without the 1,3-butanediol treatment. Pgk-1, phosphoglycerokinase-1. (D and E) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (D) and low salt versus high salt fed rats (E), for comparisons of serum IL1β. (F) Same legend as (A)–(C) for Lcn2. (G and H) ELISA data from high salt versus high salt + 1,3-butanediol treated rats (G) and low salt versus high salt fed rats (H) for comparisons of serum Lcn2; *p ≤ 0.05; **p

Figure 7.. Remission of Kidney Fibrosis, Protein…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with…

Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with βOHB
(A and B) Kidney sections of high salt-fed rats treated without (A) or with (B) 20% v/v of 1,3-butanediol were stained with Masson’s trichrome. Left: i and iv displays the images at low magnification (8×). The boxed regions in the left panel were further magnified (40×) into the right panel (ii and v). Black arrows in (ii) and (v) indicate protein casts; dark blue arrows in (iii) and (vi) denote blue staining for collagen. (C–E) Bar graphs show quantitation of renal injury (n = 4/group) (C), mRNA level of genes encoding for cellular stress and tissue fibrosis (n = 8/group) (D), and UPE (urinary protein excretion) (E). (F and G) Representative images (200× magnification) of kidney sections of high salt fed rats (F) and high salt + 1,3-butanediol fed rats (G) display immunostaining for macrophages (Cd68, brown color). Arrows indicate the infiltration of macrophages in kidney sections. (H) Quantitation of macrophage numbers. Numbers are counts of macrophages; n = 10. 10× magnification fields/group. Values are expressed as mean ± SEM (*p
All figures (7)
Similar articles
Cited by
References
    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, et al. (2012). Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol 8, e1002358. - PMC - PubMed
    1. Anders HJ, Suarez-Alvarez B, Grigorescu M, Foresto-Neto O, Steiger S, Desai J, Marschner JA, Honarpisheh M, Shi C, Jordan J, et al. (2018). The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 93, 656–669. - PubMed
    1. Arima S, Uto H, Ibusuki R, Kumamoto R, Tanoue S, Mawatari S, Oda K, Numata M, Fujita H, Oketani M, et al. (2014). Hypertension exacerbates liver injury and hepatic fibrosis induced by a choline-deficient L-amino aciddefined diet in rats. Int. J. Mol. Med 33, 68–76. - PubMed
    1. Bai M, Chen Y, Zhao M, Zhang Y, He JC, Huang S, Jia Z, and Zhang A (2017). NLRP3 inflammasome activation contributes to aldosteroneinduced podocyte injury. Am. J. Physiol. Renal Physiol 312, F556–F564. - PubMed
    1. Barbato JC, Koch LG, Darvish A, Cicila GT, Metting PJ, and Britton SL (1998). Spectrum of aerobic endurance running performance in eleven inbred strains of rats. J. Appl. Physiol. (1985) 85, 530–536. - PubMed
Show all 86 references
Publication types
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 7.. Remission of Kidney Fibrosis, Protein…
Figure 7.. Remission of Kidney Fibrosis, Protein Casts, and Urinary Proteinuria in Rats Treated with βOHB
(A and B) Kidney sections of high salt-fed rats treated without (A) or with (B) 20% v/v of 1,3-butanediol were stained with Masson’s trichrome. Left: i and iv displays the images at low magnification (8×). The boxed regions in the left panel were further magnified (40×) into the right panel (ii and v). Black arrows in (ii) and (v) indicate protein casts; dark blue arrows in (iii) and (vi) denote blue staining for collagen. (C–E) Bar graphs show quantitation of renal injury (n = 4/group) (C), mRNA level of genes encoding for cellular stress and tissue fibrosis (n = 8/group) (D), and UPE (urinary protein excretion) (E). (F and G) Representative images (200× magnification) of kidney sections of high salt fed rats (F) and high salt + 1,3-butanediol fed rats (G) display immunostaining for macrophages (Cd68, brown color). Arrows indicate the infiltration of macrophages in kidney sections. (H) Quantitation of macrophage numbers. Numbers are counts of macrophages; n = 10. 10× magnification fields/group. Values are expressed as mean ± SEM (*p
All figures (7)

References

    1. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat B, et al. (2012). Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol 8, e1002358.
    1. Anders HJ, Suarez-Alvarez B, Grigorescu M, Foresto-Neto O, Steiger S, Desai J, Marschner JA, Honarpisheh M, Shi C, Jordan J, et al. (2018). The macrophage phenotype and inflammasome component NLRP3 contributes to nephrocalcinosis-related chronic kidney disease independent from IL-1-mediated tissue injury. Kidney Int. 93, 656–669.
    1. Arima S, Uto H, Ibusuki R, Kumamoto R, Tanoue S, Mawatari S, Oda K, Numata M, Fujita H, Oketani M, et al. (2014). Hypertension exacerbates liver injury and hepatic fibrosis induced by a choline-deficient L-amino aciddefined diet in rats. Int. J. Mol. Med 33, 68–76.
    1. Bai M, Chen Y, Zhao M, Zhang Y, He JC, Huang S, Jia Z, and Zhang A (2017). NLRP3 inflammasome activation contributes to aldosteroneinduced podocyte injury. Am. J. Physiol. Renal Physiol 312, F556–F564.
    1. Barbato JC, Koch LG, Darvish A, Cicila GT, Metting PJ, and Britton SL (1998). Spectrum of aerobic endurance running performance in eleven inbred strains of rats. J. Appl. Physiol. (1985) 85, 530–536.
    1. Bomfim GF, Rodrigues FL, and Carneiro FS (2017). Are the innate and adaptive immune systems setting hypertension on fire? Pharmacol. Res 117, 377–393.
    1. Brown IJ, Tzoulaki I, Candeias V, and Elliott P (2009). Salt intakes around the world: implications for public health. Int. J. Epidemiol 38, 791–813.
    1. Bugyei-Twum A, Abadeh A, Thai K, Zhang Y, Mitchell M, Kabir G, and Connelly KA (2016). Suppression of NLRP3 inflammasome activation ameliorates chronic kidney disease-induced cardiac fibrosis and diastolic dysfunction. Sci. Rep 6, 39551.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Peña AG, Goodrich JK, Gordon JI, et al. (2010). QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336.
    1. Carpenter RG, and Grossman SP (1983). Plasma fat metabolites and hunger. Physiol. Behav 30, 57–63.
    1. Collado MC, Isolauri E, Laitinen K, and Salminen S (2008). Distinct composition of gut microbiota during pregnancy in overweight and normalweight women. Am. J. Clin. Nutr 88, 894–899.
    1. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, et al. (2016). Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 24, 256–268.
    1. Dahl LK (1961). Effects of chronic excess salt feeding. Induction of self-sustaining hypertension in rats. J. Exp. Med 114, 231–236.
    1. Dahl LK, Heine M, and Tassinari L (1964). Effects of chronic excess salt ingestion. vascular reactivity in two strains of rats with opposite genetic susceptibility to experimental hypertension. Circulation 30 (Suppl 2), 11–22.
    1. Dahl LK, Knudsen KD, and Iwai J (1970). Genetic influence of the kidney in hypertension-prone rats. Circ. Res 27 (Suppl 2), 277.
    1. De Miguel C, Das S, Lund H, and Mattson DL (2010). T lymphocytes mediate hypertension and kidney damage in Dahl salt-sensitive rats. Am. J. Physiol. Regul. Integr. Comp. Physiol 298, R1136–R1142.
    1. De Miguel C, Rudemiller NP, Abais JM, and Mattson DL (2015). Inflammation and hypertension: new understandings and potential therapeutic targets. Curr. Hypertens. Rep 17, 507.
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, Huber T, Dalevi D, Hu P, and Andersen GL (2006). Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol 72, 5069–5072.
    1. Dolinsky VW, Morton JS, Oka T, Robillard-Frayne I, Bagdan M, Lopaschuk GD, Des Rosiers C, Walsh K, Davidge ST, and Dyck JR (2010). Calorie restriction prevents hypertension and cardiac hypertrophy in the spontaneously hypertensive rat. Hypertension 56, 412–421.
    1. Edgar RC (2010). Search and clustering orders of magnitude faster than BLAST. Bioinformatics 26, 2460–2461.
    1. Evans M, Cogan KE, and Egan B (2017). Metabolism of ketone bodies during exercise and training: physiological basis for exogenous supplementation. J. Physiol 595, 2857–2871.
    1. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. (2013). Crosstalk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 110, 9066–9071.
    1. Feldman N, Rotter-Maskowitz A, and Okun E (2015). DAMPs as mediators of sterile inflammation in aging-related pathologies. Ageing Res. Rev 24 (Pt A), 29–39.
    1. Foresto-Neto O, Ávila VF, Arias SCA, Zambom FFF, Rempel LCT, Faustino VD, Machado FG, Malheiros DMAC, Abensur H, Camara NOS, et al. (2018). NLRP3 inflammasome inhibition ameliorates tubulointerstitial injury in the remnant kidney model. Lab. Invest. 98, 773–782.
    1. Garrett MR, Dene H, and Rapp JP (2003). Time-course genetic analysis of albuminuria in Dahl salt-sensitive rats on low-salt diet. J. Am. Soc. Nephrol 14, 1175–1187.
    1. Garrett MR, Joe B, and Yerga-Woolwine S (2006). Genetic linkage of urinary albumin excretion in Dahl salt-sensitive rats: influence of dietary salt and confirmation using congenic strains. Physiol. Genomics 25, 39–49.
    1. Greene AS, Yu ZY, Roman RJ, and Cowley AW Jr. (1990). Role of blood volume expansion in Dahl rat model of hypertension. Am. J. Physiol 258, H508–H514.
    1. Ha V, Sievenpiper JL, de Souza RJ, Chiavaroli L, Wang DD, Cozma AI, Mirrahimi A, Yu ME, Carleton AJ, Dibuono M, et al. (2012). Effect of fructose on blood pressure: a systematic review and meta-analysis of controlled feeding trials. Hypertension 59, 787–795.
    1. Hall SE, Wastney ME, Bolton TM, Braaten JT, and Berman M (1984). Ketone body kinetics in humans: the effects of insulin-dependent diabetes, obesity, and starvation. J. Lipid Res 25, 1184–1194.
    1. Han J, Lin K, Sequeira C, and Borchers CH (2015). An isotope-labeled chemical derivatization method for the quantitation of short-chain fatty acids in human feces by liquid chromatography-tandem mass spectrometry. Anal. Chim. Acta 854, 86–94.
    1. Hattori T, Murase T, Takatsu M, Nagasawa K, Matsuura N, Watanabe S, Murohara T, and Nagata K (2014). Dietary salt restriction improves cardiac and adipose tissue pathology independently of obesity in a rat model of metabolic syndrome. J. Am. Heart Assoc 3, e001312.
    1. Huang B, Cheng Y, Usa K, Liu Y, Baker MA, Mattson DL, He Y, Wang N, and Liang M (2016). Renal Tumor Necrosis Factor α Contributes to Hypertension in Dahl Salt-Sensitive Rats. Sci. Rep 6, 21960.
    1. Hutton HL, Ooi JD, Holdsworth SR, and Kitching AR (2016). The NLRP3 inflammasome in kidney disease and autoimmunity. Nephrology (Carlton) 21, 736–744.
    1. Karlsson CL, Onnerfält J, Xu J, Molin G, Ahrné S, and Thorngren-Jerneck K (2012). The microbiota of the gut in preschool children with normal and excessive body weight. Obesity (Silver Spring) 20, 2257–2261.
    1. Kersse K, Bertrand MJ, Lamkanfi M, and Vandenabeele P (2011). NODlike receptors and the innate immune system: coping with danger, damage and death. Cytokine Growth Factor Rev. 22, 257–276.
    1. Kimura T, Hashimoto Y, Tanaka M, Asano M, Yamazaki M, Oda Y, Toda H, Marunaka Y, Nakamura N, and Fukui M (2016). Sodium-chloride difference and metabolic syndrome: a population-based large-scale cohort study. Intern. Med 55, 3085–3090.
    1. Kitada K, Daub S, Zhang Y, Klein JD, Nakano D, Pedchenko T, Lantier L, LaRocque LM, Marton A, Neubert P, et al. (2017). High salt intake reprioritizes osmolyte and energy metabolism for body fluid conservation. J. Clin. Invest 127, 1944–1959.
    1. Kumarasamy S, Gopalakrishnan K, Abdul-Majeed S, Partow-Navid R, Farms P, and Joe B (2013). Construction of two novel reciprocal conplastic rat strains and characterization of cardiac mitochondria. Am. J. Physiol. Heart Circ. Physiol 304, H22–H32.
    1. Lanaspa MA, Kuwabara M, Andres-Hernando A, Li N, Cicerchi C, Jensen T, Orlicky DJ, Roncal-Jimenez CA, Ishimoto T, Nakagawa T, et al. (2018). High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism. Proc. Natl. Acad. Sci. USA 115, 3138–3143.
    1. Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, et al. (2013). Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat. Biotechnol 31, 814–821.
    1. Lemos DR, McMurdo M, Karaca G, Wilflingseder J, Leaf IA, Gupta N, Miyoshi T, Susa K, Johnson BG, Soliman K, et al. (2018). Interleukin-1b activates a MYC-dependent metabolic switch in kidney stromal cells necessary for progressive tubulointerstitial fibrosis. J. Am. Soc. Nephrol 29, 1690–1705.
    1. Livak KJ, and Schmittgen TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25, 402–408.
    1. Maalouf M, Rho JM, and Mattson MP (2009). The neuroprotective properties of calorie restriction, the ketogenic diet, and ketone bodies. Brain Res. Brain Res. Rev 59, 293–315.
    1. Mahoney LB, Denny CA, and Seyfried TN (2006). Caloric restriction in C57BL/6J mice mimics therapeutic fasting in humans. Lipids Health Dis. 5, 13.
    1. Mattson DL (2014). Infiltrating immune cells in the kidney in salt-sensitive hypertension and renal injury. Am. J. Physiol. Renal Physiol 307, F499–F508.
    1. Mattson DL, James L, Berdan EA, and Meister CJ (2006). Immune suppression attenuates hypertension and renal disease in the Dahl salt-sensitive rat. Hypertension 48, 149–156.
    1. Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, Vijay-Kumar M, Pennathur S, and Joe B (2015). Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol. Genomics 47, 187–197.
    1. Melo DS, Costa-Pereira LV, Santos CS, Mendes BF, Costa KB, Santos CF, Rocha-Vieira E, Magalhães FC, Esteves EA, Ferreira AJ, et al. (2016). Severe calorie restriction reduces cardiometabolic risk factors and protects rat hearts from ischemia/reperfusion injury. Front. Physiol 7, 106.
    1. Musso G, Gambino R, De Michieli F, Cassader M, Rizzetto M, Durazzo M, Fagà E, Silli B, and Pagano G (2003). Dietary habits and their relations to insulin resistance and postprandial lipemia in nonalcoholic steatohepatitis. Hepatology 37, 909–916.
    1. Newman JC, and Verdin E (2014). β-hydroxybutyrate: much more than a metabolite. Diabetes Res. Clin. Pract 106, 173–181.
    1. Newman JC, and Verdin E (2017). β-hydroxybutyrate: a signaling metabolite. Annu. Rev. Nutr 37, 51–76.
    1. Padmanabhan S, and Joe B (2017). Towards precision medicine for hypertension: a review of genomic, epigenomic, and microbiomic effects on blood pressure in experimental rat models and humans. Physiol. Rev 97, 1469–1528.
    1. Paragas N, Qiu A, Zhang Q, Samstein B, Deng SX, Schmidt-Ott KM, Viltard M, Yu W, Forster CS, Gong G, et al. (2011). The Ngal reporter mouse detects the response of the kidney to injury in real time. Nat. Med 17, 216–222.
    1. Puchalska P, and Crawford PA (2017). Multi-dimensional Roles of Ketone Bodies in Fuel Metabolism, Signaling, and Therapeutics. Cell Metab. 25, 262–284.
    1. Rapp JP, and Dene H (1985). Development and characteristics of inbred strains of Dahl salt-sensitive and salt-resistant rats. Hypertension 7, 340–349.
    1. Rapp JP, Knudsen KD, Iwai J, and Dahl LK (1973). Genetic control of blood pressure and corticosteroid production in rats. Circ. Res 32 (Suppl 1), 139–149.
    1. Ringseis R, Eder K, Mooren FC, and Kru€ger K (2015). Metabolic signals and innate immune activation in obesity and exercise. Exerc. Immunol. Rev 21, 58–68.
    1. Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, Perez G, Gutierrez-Casado E, Koike S, Knotts TA, et al. (2017). A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 26, 539–546.
    1. Rossi R, Dörig S, Del Prete E, and Scharrer E (2000). Suppression of feed intake after parenteral administration of D-beta-hydroxybutyrate in pygmy goats. J. Vet. Med. A Physiol. Pathol. Clin. Med 47, 9–16.
    1. Rudemiller N, Lund H, Jacob HJ, Geurts AM, and Mattson DL; PhysGen Knockout Program (2014). CD247 modulates blood pressure by altering T-lymphocyte infiltration in the kidney. Hypertension 63, 559–564.
    1. Santacruz A, Collado MC, García-Valdés L, Segura MT, Martín-Lagos JA, Anjos T, Martí-Romero M, Lopez RM, Florido J, Campoy C, and Sanz Y (2010). Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr 104, 83–92.
    1. Savage CD, Lopez-Castejon G, Denes A, and Brough D (2012). NLRP3-inflammasome activating DAMPs stimulate an inflammatory response in glia in the absence of priming which contributes to brain inflammation after injury. Front. Immunol 3, 288.
    1. Schneeberger M, Everard A, Gómez-Valadés AG, Matamoros S, Ramírez S, Delzenne NM, Gomis R, Claret M, and Cani PD (2015). Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci. Rep 5, 16643.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, and Huttenhower C (2011). Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60.
    1. Sepehri Z, Kiani Z, Afshari M, Kohan F, Dalvand A, and Ghavami S (2017). Inflammasomes and type 2 diabetes: An updated systematic review. Immunol. Lett 192, 97–103.
    1. Stienstra R, van Diepen JA, Tack CJ, Zaki MH, van de Veerdonk FL, Perera D, Neale GA, Hooiveld GJ, Hijmans A, Vroegrijk I, et al. (2011). Inflammasome is a central player in the induction of obesity and insulin resistance. Proc. Natl. Acad. Sci. USA 108, 15324–15329.
    1. Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, and Peterson CM (2018). Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 27, 1212–1221.
    1. Theriot CM, Koenigsknecht MJ, Carlson PE Jr., Hatton GE, Nelson AM, Li B, Huffnagle GB, Z Li J, and Young VB (2014). Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat. Commun 5, 3114.
    1. Tian N, Moore RS, Braddy S, Rose RA, Gu JW, Hughson MD, and Manning RD Jr. (2007). Interactions between oxidative stress and inflammation in salt-sensitive hypertension. Am. J. Physiol. Heart Circ. Physiol 293, H3388–H3395.
    1. Uetake Y, Ikeda H, Irie R, Tejima K, Matsui H, Ogura S, Wang H, Mu S, Hirohama D, Ando K, et al. (2015). High-salt in addition to high-fat diet may enhance inflammation and fibrosis in liver steatosis induced by oxidative stress and dyslipidemia in mice. Lipids Health Dis. 14, 6.
    1. Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM, and Dixit VD (2011). The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med 17, 179–188.
    1. Veech RL, Bradshaw PC, Clarke K, Curtis W, Pawlosky R, and King MT (2017). Ketone bodies mimic the life span extending properties of caloric restriction. IUBMB Life 69, 305–314.
    1. Vilaysane A, Chun J, Seamone ME, Wang W, Chin R, Hirota S, Li Y, Clark SA, Tschopp J, Trpkov K, et al. (2010). The NLRP3 inflammasome promotes renal inflammation and contributes to CKD. J. Am. Soc. Nephrol 21, 1732–1744.
    1. Wan Z, Wen W, Ren K, Zhou D, Liu J, Wu Y, Zhou J, Mu J, and Yuan Z (2018). Involvement of NLRP3 inflammasome in the impacts of sodium and potassium on insulin resistance in normotensive Asians. Br. J. Nutr 119, 228–237.
    1. Wang G, Yeung CK, Wong WY, Zhang N, Wei YF, Zhang JL, Yan Y, Wong CY, Tang JJ, Chuai M, et al. (2016a). Liver fibrosis can be induced by high salt intake through excess reactive oxygen species (ROS) production. J. Agric. Food Chem 64, 1610–1617.
    1. Wang Y, Xu Y, Sheng H, Ni X, and Lu J (2016b). Exercise amelioration of depression-like behavior in OVX mice is associated with suppression of NLRP3 inflammasome activation in hippocampus. Behav. Brain Res 307, 18–24.
    1. Wang X, Liu Q, Zhou J, Wu X, and Zhu Q (2017). b hydroxybutyrate levels in serum and cerebrospinal fluid under ketone body metabolism in rats. Exp. Anim 66, 177–182.
    1. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, Haase S, Mähler A, Balogh A, Markó L, et al. (2017). Salt-responsive gut commensal modulates TH17 axis and disease. Nature 551, 585–589.
    1. Willner IR, Waters B, Patil SR, Reuben A, Morelli J, and Riely CA (2001). Ninety patients with nonalcoholic steatohepatitis: insulin resistance, familial tendency, and severity of disease. Am. J. Gastroenterol 96, 2957–2961.
    1. World Health Organization (2003). Diet, nutrition and the prevention of chronic diseases. WHO Tech. Rep. Ser 916, 1–149.
    1. Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, Regev A, and Kuchroo VK (2013). Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature 496, 513–517.
    1. Xia J, Psychogios N, Young N, and Wishart DS (2009). MetaboAnalyst: a web server for metabolomic data analysis and interpretation. Nucleic Acids Res. 37, W652–60.
    1. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’Agostino D, Planavsky N, Lupfer C, Kanneganti TD, et al. (2015). The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med 21, 263–269.
    1. Yu J, Nagasu H, Murakami T, Hoang H, Broderick L, Hoffman HM, and Horng T (2014). Inflammasome activation leads to Caspase-1-dependent mitochondrial damage and block of mitophagy. Proc. Natl. Acad. Sci. USA 111, 15514–15519.
    1. Zhu Q, Li XX, Wang W, Hu J, Li PL, Conley S, and Li N (2016). Mesenchymal stem cell transplantation inhibited high salt-induced activation of the NLRP3 inflammasome in the renal medulla in Dahl S rats. Am. J. Physiol. Renal Physiol 310, F621–F627.
    1. Ziegler TR, Luo M, Estívariz CF, Moore DA 3rd, Sitaraman SV, Hao L, Bazargan N, Klapproth JM, Tian J, Galloway JR, et al. (2008). Detectable serum flagellin and lipopolysaccharide and upregulated anti-flagellin and lipopolysaccharide immunoglobulins in human short bowel syndrome. Am. J. Physiol. Regul. Integr. Comp. Physiol 294, R402–R410.

Source: PubMed

3
Prenumerera