Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38779751

ABSTRACT

Sepsis-associated acute kidney injury (SA-AKI) is a key contributor to the life threatening sequalae of sepsis. Mechanistically, SA-AKI is a consequence of unabated myeloid cell activation and oxidative stress that induces tubular injury. Iron mediates inflammatory pathways directly and through regulating the expression of ferritin, an iron storage protein comprised of ferritin light (FtL) and heavy chain (FtH). Previous work revealed myeloid FtH deletion leads to a compensatory increase in intracellular and circulating FtL and is associated with amelioration of SA-AKI. We designed this study to test the hypothesis that loss of myeloid FtL will exacerbate the sepsis-induced inflammatory response and worsen SA-AKI. We generated a novel myeloid-specific FtL knockout mouse and induced sepsis via cecal ligation and puncture or lipopolysaccharide endotoxemia. As expected, myeloid FtL and serum ferritin levels were significantly lower in the knockout mice. Interestingly, while sepsis led to production of pro- and anti-inflammatory cytokines, there was no statistical difference between the genotypes. There was a similar loss of kidney function and injury, identified by expression of kidney injury molecule-1 and neutrophil gelatinase associated lipocalin. RNA sequencing revealed upregulation of pathways for cell cycle arrest and autophagy post-sepsis, but no significant differences were observed between genotypes, including in key genes associated with ferroptosis, an iron-mediated form of cell death. FtL deletion did not impact activation of NFkB or HIF-1a signaling, key inflammatory pathways associated with dysregulated host response. Taken together, while FtL overexpression was shown to be protective, loss of FtL did not influence sepsis pathogenesis.

2.
Kidney Int ; 105(5): 971-979, 2024 May.
Article in English | MEDLINE | ID: mdl-38290599

ABSTRACT

Demand for kidney grafts outpaces supply, limiting kidney transplantation as a treatment for kidney failure. Xenotransplantation has the potential to make kidney transplantation available to many more patients with kidney failure, but the ability of xenografts to support human physiologic homeostasis has not been established. A brain-dead adult decedent underwent bilateral native nephrectomies followed by 10 gene-edited (four gene knockouts, six human transgenes) pig-to-human xenotransplantation. Physiologic parameters and laboratory values were measured for seven days in a critical care setting. Data collection aimed to assess homeostasis by measuring components of the renin-angiotensin-aldosterone system, parathyroid hormone signaling, glomerular filtration rate, and markers of salt and water balance. Mean arterial blood pressure was maintained above 60 mmHg throughout. Pig kidneys secreted renin (post-operative day three to seven mean and standard deviation: 47.3 ± 9 pg/mL). Aldosterone and angiotensin II levels were present (post-operative day three to seven, 57.0 ± 8 pg/mL and 5.4 ± 4.3 pg/mL, respectively) despite plasma renin activity under 0.6 ng/mL/hr. Parathyroid hormone levels followed ionized calcium. Urine output down trended from 37 L to 6 L per day with 4.5 L of electrolyte free water loss on post-operative day six. Aquaporin 2 channels were detected in the apical surface of principal cells, supporting pig kidney response to human vasopressin. Serum creatinine down trended to 0.9 mg/dL by day seven. Glomerular filtration rate ranged 90-240 mL/min by creatinine clearance and single-dose inulin clearance. Thus, in a human decedent model, xenotransplantation of 10 gene-edited pig kidneys provided physiologic balance for seven days. Hence, our in-human study paves the way for future clinical study of pig-to-human kidney xenotransplantation in living persons.


Subject(s)
Renal Insufficiency , Renin , Adult , Humans , Animals , Swine , Transplantation, Heterologous , Kidney/physiology , Renin-Angiotensin System , Aldosterone , Homeostasis , Parathyroid Hormone , Water
3.
Am J Physiol Renal Physiol ; 325(6): F717-F732, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37767569

ABSTRACT

Daily, we may experience mild dehydration with a rise in plasma osmolality that triggers the release of vasopressin. Although the effect of dehydration is well characterized in collecting duct principal cells (CDPCs), we hypothesized that mild dehydration (<12 h) results in many kidney cell-specific changes in transcriptomes and chromatin accessibility. Single-nucleus (sn) multiome (RNA-assay for transposase-accessible chromatin) sequencing and bulk RNA sequencing of kidneys from male and female mice that were mildly water deprived or not were compared. Water-deprived mice had a significant increase in plasma osmolality. sn-multiome-seq resulted in 19,837 nuclei that were annotated into 33 clusters. In CDPCs, aquaporin 2 (Aqp2) and aquaporin 3 (Apq3) were greater in dehydrated mice, but there were novel genes like gremlin 2 (Grem2; a cytokine) that were increased compared with ad libitum mice. The transcription factor cAMP-responsive element modulator (Crem) was greater in CDPCs of dehydrated mice, and the Crem DNA motif was more accessible. There were hundreds of sex- and dehydration-specific differentially expressed genes (DEGs) throughout the kidney, especially in the proximal tubules and thin limbs. In male mice, DEGs were enriched in pathways related to lipid metabolism, whereas female DEGs were enriched in organic acid metabolism. Many highly expressed genes had a positive correlation with increased chromatin accessibility, and mild dehydration exerted many transcriptional changes that we detected at the chromatin level. Even with a rise in plasma osmolality, male and female kidneys have distinct transcriptomes suggesting that there may be diverse mechanisms used to remain in fluid balance.NEW & NOTEWORTHY The kidney consists of >30 cell types that work collectively to maintain fluid-electrolyte balance. Kidney single-nucleus transcriptomes and chromatin accessibility profiles from male and female control (ad libitum water and food) or mildly dehydrated mice (ad libitum food, water deprivation) were determined. Mild dehydration caused hundreds of cell- and sex-specific transcriptomic changes, even though the kidney function to conserve water was the same.


Subject(s)
Dehydration , Transcriptome , Mice , Animals , Male , Female , Dehydration/metabolism , Chromatin/genetics , Chromatin/metabolism , Aquaporin 2/genetics , Aquaporin 2/metabolism , Kidney/metabolism , Water/metabolism
5.
Article in English | MEDLINE | ID: mdl-36169157

ABSTRACT

The Seventeenth International Conference on Endothelin (ET-17) was held during 4-7 October 2021 and because of the SARS-CoV-2 pandemic it was held virtually. Sponsored by the American Physiological Society, ET-17 was held over 4 half-days, with exciting studies related to all organ systems presented. Since the Lancet article reporting the successful SONAR clinical trial with endothelin receptor A blockade in diabetic nephropathy, there has been renewed interest in the use of endothelin receptor antagonists in the treatment of a variety of diseases. From the rigorous preclinical studies to the latest clinical trials, ET-17 was full of exciting science, some of which is reported in this special issue. We welcomed new labs to the meeting and everyone left with the impression that ET-related research is a vibrant field with very significant discoveries being made.

6.
Can J Physiol Pharmacol ; 100(9): 868-879, 2022 Sep 01.
Article in English | MEDLINE | ID: mdl-35704945

ABSTRACT

The chemotherapeutic agent cisplatin accumulates in the kidney and induces acute kidney injury (AKI). Preclinical and clinical studies suggest that young female mice and women show greater recovery from cisplatin-AKI compared to young male mice and men. The endothelin (ET) and ET receptors are enriched in the kidney and may be dysfunctional in cisplatin-AKI; however, there is a gap in our knowledge about the putative effects of sex and cisplatin on the renal ET system. We hypothesized that cisplatin-AKI male and female mice will have increased expression of the renal ET system. As expected, all cisplatin-AKI mice had kidney damage and body weight loss greater than control mice. Cisplatin-AKI mice had greater cortical Edn1, Edn3, Ednra, and Ednrb, while outer medullary Ednra was significantly suppressed in both sexes. Of the ∼25 000 genes sequenced from the inner medulla, only 91 genes (comparing saline mice) and 134 genes (comparing cisplatin-AKI mice) were differentially expressed and they were unrelated to the ET system. However, Edn1 was significantly greater in the inner medulla of male and female cisplatin-AKI mice. Thus, RNA profiles of the ET system were significantly affected by cisplatin-AKI throughout the kidney regardless of sex and this may help determine the therapeutic potential of targeting the ET receptors in cisplatin-AKI.


Subject(s)
Acute Kidney Injury , Antineoplastic Agents , Cisplatin , Endothelin-1 , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/genetics , Animals , Antineoplastic Agents/toxicity , Apoptosis , Cisplatin/toxicity , Endothelin-1/metabolism , Female , Kidney , Male , Mice , Mice, Inbred C57BL
7.
J Am Heart Assoc ; 11(5): e020450, 2022 03.
Article in English | MEDLINE | ID: mdl-35191321

ABSTRACT

Background Premenopausal women are less likely to develop hypertension and salt-related complications than are men, yet the impact of sex on mechanisms regulating Na+ homeostasis during dietary salt challenges is poorly defined. Here, we determined whether female rats have a more efficient capacity to acclimate to increased dietary salt intake challenge. Methods and Results Age-matched male and female Sprague Dawley rats maintained on a normal-salt (NS) diet (0.49% NaCl) were challenged with a 5-day high-salt diet (4.0% NaCl). We assessed serum, urinary, skin, and muscle electrolytes; total body water; and kidney Na+ transporters during the NS and high-salt diet phases. During the 5-day high-salt challenge, natriuresis increased more rapidly in females, whereas serum Na+ and body water concentration increased only in males. To determine if females are primed to handle changes in dietary salt, we asked the question whether the renal endothelin-1 natriuretic system is more active in female rats, compared with males. During the NS diet, female rats had a higher urinary endothelin-1 excretion rate than males. Moreover, Ingenuity Pathway Analysis of RNA sequencing data identified the enrichment of endothelin signaling pathway transcripts in the inner medulla of kidneys from NS-fed female rats compared with male counterparts. Notably, in human subjects who consumed an Na+-controlled diet (3314-3668 mg/day) for 3 days, women had a higher urinary endothelin-1 excretion rate than men, consistent with our findings in NS-fed rats. Conclusions These results suggest that female sex confers a greater ability to maintain Na+ homeostasis during acclimation to dietary Na+ challenges and indicate that the intrarenal endothelin-1 natriuretic pathway is enhanced in women.


Subject(s)
Sodium Chloride, Dietary , Sodium Chloride , Acclimatization , Animals , Blood Pressure , Diet , Endothelin-1/metabolism , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Sodium , Sodium Chloride, Dietary/metabolism
8.
J Am Soc Nephrol ; 33(4): 769-785, 2022 04.
Article in English | MEDLINE | ID: mdl-35115326

ABSTRACT

BACKGROUND: Vascular congestion of the renal medulla-trapped red blood cells in the medullary microvasculature-is a hallmark finding at autopsy in patients with ischemic acute tubular necrosis. Despite this, the pathogenesis of vascular congestion is not well defined. METHODS: In this study, to investigate the pathogenesis of vascular congestion and its role in promoting renal injury, we assessed renal vascular congestion and tubular injury after ischemia reperfusion in rats pretreated with low-dose LPS or saline (control). We used laser Doppler flowmetry to determine whether pretreatment with low-dose LPS prevented vascular congestion by altering renal hemodynamics during reperfusion. RESULTS: We found that vascular congestion originated during the ischemic period in the renal venous circulation. In control animals, the return of blood flow was followed by the development of congestion in the capillary plexus of the outer medulla and severe tubular injury early in reperfusion. Laser Doppler flowmetry indicated that blood flow returned rapidly to the medulla, several minutes before recovery of full cortical perfusion. In contrast, LPS pretreatment prevented both the formation of medullary congestion and its associated tubular injury. Laser Doppler flowmetry in LPS-pretreated rats suggested that limiting early reperfusion of the medulla facilitated this protective effect, because it allowed cortical perfusion to recover and clear congestion from the large cortical veins, which also drain the medulla. CONCLUSIONS: Blockage of the renal venous vessels and a mismatch in the timing of cortical and medullary reperfusion results in congestion of the outer medulla's capillary plexus and promotes early tubular injury after renal ischemia. These findings indicate that hemodynamics during reperfusion contribute to the renal medulla's susceptibility to ischemic injury.


Subject(s)
Acute Kidney Injury , Reperfusion Injury , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Humans , Ischemia/complications , Kidney/pathology , Kidney Medulla/blood supply , Lipopolysaccharides , Rats , Renal Circulation/physiology , Reperfusion/adverse effects , Reperfusion Injury/complications , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control
9.
Physiol Genomics ; 54(2): 45-57, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34890513

ABSTRACT

Recent studies have identified at least 20 different kidney cell types based upon chromatin structure and gene expression. Histone deacetylases (HDACs) are epigenetic transcriptional repressors via deacetylation of histone lysines resulting in inaccessible chromatin. We reported that kidney epithelial HDAC1 and HDAC2 activity is critical for maintaining a healthy kidney and preventing fluid-electrolyte abnormalities. However, to what extent does Hdac1/Hdac2 knockdown affect chromatin structure and subsequent transcript expression in the kidney? To answer this question, we used single nucleus assay for transposase-accessible chromatin-sequencing (snATAC-seq) and snRNA-seq to profile kidney nuclei from male and female, control, and littermate kidney epithelial Hdac1/Hdac2 knockdown mice. Hdac1/Hdac2 knockdown resulted in significant changes in the chromatin structure predominantly within the promoter region of gene loci involved in fluid-electrolyte balance such as the aquaporins, with both increased and decreased accessibility captured. Moreover, Hdac1/Hdac2 knockdown resulted different gene loci being accessible with a corresponding increased transcript number in the kidney, but among all mice only 24%-30% of chromatin accessibility agreed with transcript expression (e.g., open chromatin and increased transcript). To conclude, although chromatin structure does affect transcription, ∼70% of the differentially expressed genes cannot be explained by changes in chromatin accessibility and HDAC1/HDAC2 had a minimal effect on these global patterns. Yet, the genes that are targets of HDAC1 and HDAC2 are critically important for maintaining kidney function.


Subject(s)
Chromatin/genetics , Epithelial Cells/metabolism , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Kidney/metabolism , Transcriptome/genetics , Animals , Aquaporin 1/genetics , Aquaporin 1/metabolism , Aquaporin 2/genetics , Aquaporin 2/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing/methods , Female , Gene Expression Profiling/methods , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Kidney/cytology , Male , Mice, Knockout , RNA-Seq/methods
10.
Physiol Rep ; 9(20): e15080, 2021 10.
Article in English | MEDLINE | ID: mdl-34665521

ABSTRACT

The nitric oxide (NO)-generating enzyme, NO synthase-1ß (NOS1ß), is essential for sodium (Na+ ) homeostasis and blood pressure control. We previously showed that collecting duct principal cell NOS1ß is critical for inhibition of the epithelial sodium channel (ENaC) during high Na+ intake. Previous studies on freshly isolated cortical collecting ducts (CCD) demonstrated that exogenous NO promotes basolateral potassium (K+ ) conductance through basolateral channels, presumably Kir 4.1 (Kcnj10) and Kir 5.1 (Kcnj16). We, therefore, investigated the effects of NOS1ß knockout on Kir 4.1/Kir 5.1 channel activity. Indeed, in CHO cells overexpressing NOS1ß and Kir 4.1/Kir 5.1, the inhibition of NO signaling decreased channel activity. Male littermate control and principal cell NOS1ß knockout mice (CDNOS1KO) on a 7-day, 4% NaCl diet (HSD) were used to detect changes in basolateral K+ conductance. We previously demonstrated that CDNOS1KO mice have high circulating aldosterone despite a high-salt diet and appropriately suppressed renin. We observed greater Kir 4.1 cortical abundance and significantly greater Kir 4.1/Kir 5.1 single-channel activity in the principal cells from CDNOS1KO mice. Moreover, blocking aldosterone action with in vivo spironolactone treatment resulted in lower Kir 4.1 abundance and greater plasma K+ in the CDNOS1KO mice compared to controls. Lowering K+ content in the HSD prevented the high aldosterone and greater plasma Na+ of CDNOS1KO mice and normalized Kir 4.1 abundance. We conclude that during chronic HSD, lack of NOS1ß leads to increased plasma K+ , enhanced circulating aldosterone, and activation of ENaC and Kir 4.1/Kir 5.1 channels. Thus, principal cell NOS1ß is required for the regulation of both Na+ and K+ by the kidney.


Subject(s)
Homeostasis , Kidney Tubules, Collecting/metabolism , Nitric Oxide Synthase Type I/physiology , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Sodium/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Ion Transport , Male , Mice , Mice, Knockout , Potassium Channels, Inwardly Rectifying/genetics
11.
Physiol Rep ; 9(10): e14844, 2021 05.
Article in English | MEDLINE | ID: mdl-34042301

ABSTRACT

Early life stress (ELS) is associated with cardiovascular disease (CVD) risk in adulthood, but the underlying vascular mechanisms are poorly understood. Increased hemoglobin and heme have recently been implicated to mediate endothelial dysfunction in several vascular diseases. Chronic physiological stress is associated with alterations in the heme pathway that have been well-described in the literature. However, very little is known about the heme pathway with exposure to ELS or chronic psychosocial stress. Utilizing a mouse model of ELS, maternal separation with early weaning (MSEW), we previously reported that MSEW induces endothelial dysfunction via increased superoxide production. We reasoned that heme dysregulation may be one of the culprits induced by MSEW and sustained throughout adulthood; thus, we hypothesized that MSEW induces heme dysfunction. We investigated whether circulating levels of heme, a circulating pro-oxidant mediator, are increased by MSEW and examined the role of the heme metabolic pathway and heme homeostasis in this process. We found that circulating levels of heme are increased in mice exposed to MSEW and that plasma from MSEW mice stimulated higher superoxide production in cultured mouse aortic endothelial cells (MAECs) compared to plasma from normally reared mice. The heme scavenger hemopexin blunted this enhanced superoxide production. Splenic haptoglobin abundance was significantly lower and hemoglobin levels per red blood cell were significantly higher in MSEW versus control mice. These findings lead us to propose that ELS induces increased circulating heme through dysregulation of the haptoglobin-hemoglobin system representing a mechanistic link between ELS and CVD risk in adulthood.


Subject(s)
Heme/metabolism , Maternal Deprivation , Signal Transduction/physiology , Stress, Psychological/blood , Stress, Psychological/psychology , Weaning , Age Factors , Animals , Animals, Newborn , Endothelium, Vascular/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy
12.
J Am Soc Nephrol ; 32(9): 2210-2222, 2021 09.
Article in English | MEDLINE | ID: mdl-34045314

ABSTRACT

BACKGROUND: Primary cilia regulation of renal function and BP in health and disease is incompletely understood. This study investigated the effect of nephron ciliary loss on renal physiology, BP, and ensuing cystogenesis. METHODS: Mice underwent doxycycline (DOX)-inducible nephron-specific knockout (KO) of the Ift88 gene at 2 months of age using a Cre-LoxP strategy. BP, kidney function, and renal pathology were studied 2 and 9 months after DOX (Ift88 KO) or vehicle (control). RESULTS: At 2 months post-DOX, male, but not female, Ift88 KO, compared with sex-matched control, mice had reduced BP, enhanced salt-induced natriuresis, increased urinary nitrite and nitrate (NOx) excretion, and increased kidney NOS3 levels, which localized to the outer medulla; the reductions in BP in male mice were prevented by L-NAME. At 9 months post-DOX, male, but not female, Ift88 KO mice had polycystic kidneys, elevated BP, and reduced urinary NOx excretion. No differences were observed in plasma renin concentration, plasma aldosterone, urine vasopressin, or urine PGE2 between Ift88 KO and control mice at 2 or 9 months post-DOX. CONCLUSIONS: Nephron cilia disruption in male, but not female, mice (1) reduces BP prior to cyst formation, (2) increases NOx production that may account for the lower BP prior to cyst formation, and (3) induces polycystic kidneys that are associated with hypertension and reduced renal NO production.


Subject(s)
Blood Pressure/physiology , Nephrons/physiopathology , Polycystic Kidney Diseases/etiology , Tumor Suppressor Proteins/genetics , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Knockout , Natriuresis , Nitrates/urine , Nitric Oxide Synthase Type III/metabolism , Nitrites/urine , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Sex Factors
14.
Am J Physiol Renal Physiol ; 320(3): F297-F307, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33356953

ABSTRACT

We reported that high salt (HS) intake stimulates renal collecting duct (CD) endothelin (ET) type B receptor (ETBR)/nitric oxide (NO) synthase 1ß (NOS1ß)-dependent NO production inhibiting the epithelial sodium channel (ENaC) promoting natriuresis. However, the mechanism underlying the HS-induced increase of NO production is unclear. Histone deacetylase 1 (HDAC1) responds to increased fluid flow, as can occur in the CD during HS intake. The renal inner medulla (IM), in particular the IMCD, has the highest NOS1 activity within the kidney. Hence, we hypothesized that HS intake provokes HDAC1 activation of NO production in the IM. HS intake for 1 wk significantly increased HDAC1 abundance in the IM. Ex vivo treatment of dissociated IM from HS-fed mice with a selective HDAC1 inhibitor (MS-275) decreased NO production with no change in ET-1 peptide or mRNA levels. We further investigated the role of the ET-1/ETBR/NOS1ß signaling pathway with chronic ETBR blockade (A-192621). Although NO was decreased and ET-1 levels were elevated in the dissociated IM from HS-fed mice treated with A-192621, ex vivo MS-275 did not further change NO or ET-1 levels suggesting that HDAC1-mediated NO production is regulated at the level or downstream of ETBR activation. In split-open CDs from HS-fed mice, patch clamp analysis revealed significantly higher ENaC activity after MS-275 pretreatment, which was abrogated by an exogenous NO donor. Moreover, flow-induced increases in mIMCD-3 cell NO production were blunted by HDAC1 or calcium inhibition. Taken together, these findings indicate that HS intake induces HDAC1-dependent activation of the ETBR/NO pathway contributing to the natriuretic response.


Subject(s)
Histone Deacetylase 1/metabolism , Kidney Tubules, Collecting/enzymology , Natriuresis , Nitric Oxide/metabolism , Renal Elimination , Sodium Chloride, Dietary/administration & dosage , Animals , Endothelin-1/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type I/metabolism , Receptor, Endothelin B/metabolism , Signal Transduction , Sodium Chloride, Dietary/urine
15.
JCI Insight ; 5(16)2020 08 20.
Article in English | MEDLINE | ID: mdl-32673289

ABSTRACT

Histone deacetylase (HDAC) enzymes regulate transcription through epigenetic modification of chromatin structure, but their specific functions in the kidney remain elusive. We discovered that the human kidney expresses class I HDACs. Kidney medulla-specific inhibition of class I HDACs in the rat during high-salt feeding results in hypertension, polyuria, hypokalemia, and nitric oxide deficiency. Three new inducible murine models were used to determine that HDAC1 and HDAC2 in the kidney epithelium are necessary for maintaining epithelial integrity and maintaining fluid-electrolyte balance during increased dietary sodium intake. Moreover, single-nucleus RNA-sequencing determined that epithelial HDAC1 and HDAC2 are necessary for expression of many sodium or water transporters and channels. In performing a systematic review and meta-analysis of serious adverse events associated with clinical HDAC inhibitor use, we found that HDAC inhibitors increased the odds ratio of experiencing fluid-electrolyte disorders, such as hypokalemia. This study provides insight on the mechanisms of potential serious adverse events with HDAC inhibitors, which may be fatal to critically ill patients. In conclusion, kidney tubular HDACs provide a link between the environment, such as consumption of high-salt diets, and regulation of homeostatic mechanisms to remain in fluid-electrolyte balance.


Subject(s)
Electrolytes/metabolism , Histone Deacetylase Inhibitors/adverse effects , Histone Deacetylases/metabolism , Kidney Medulla/metabolism , Animals , Benzamides/pharmacology , Blood Pressure/drug effects , Female , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Homeostasis/drug effects , Homeostasis/physiology , Humans , Kidney Medulla/drug effects , Kidney Medulla/physiopathology , Male , Nitric Oxide/metabolism , Potassium/blood , Pyridines/pharmacology , Rats, Sprague-Dawley , Sodium Chloride, Dietary/pharmacology , Water-Electrolyte Balance/physiology
16.
Hypertension ; 75(6): 1624-1634, 2020 06.
Article in English | MEDLINE | ID: mdl-32306766

ABSTRACT

The diurnal rhythms of sodium handling and blood pressure are thought to be regulated by clock genes, such as Bmal1. However, little is known about the regulation of these factors by Bmal1, especially in rats. Using a novel whole-body Bmal1 knockout rat model (Bmal1-/-), we hypothesized that time of day regulation of sodium excretion is dependent on Bmal1. Using telemetry to continuously record mean arterial pressure, we observed that male and female Bmal1-/- rats had significantly reduced mean arterial pressure over the course of 24 hours compared with littermate controls. The circadian mean arterial pressure pattern remained intact in both sexes of Bmal1-/- rats, which is in contrast to the Bmal1-/- mouse model. Male Bmal1-/- rats had no significant difference in baseline sodium excretion between 12-hour active and inactive periods, indicating a lack of diurnal control independent of maintained mean arterial pressure rhythms. Female Bmal1-/- rats, however, had significantly greater sodium excretion during the active versus inactive period similar to controls. Thus, we observed a clear dissociation between circadian blood pressure and control of sodium excretion that is sex dependent. These findings are consistent with a more robust ability of females to maintain control of sodium excretion, and furthermore, demonstrate a novel role for Bmal1 in control of diurnal blood pressure independent of sodium excretion.


Subject(s)
ARNTL Transcription Factors/metabolism , Circadian Rhythm/physiology , Kidney , Renal Elimination/physiology , Sodium/metabolism , Animals , Animals, Genetically Modified , Blood Pressure/physiology , Female , Kidney/metabolism , Kidney/physiopathology , Male , Mice , Rats , Sex Factors
17.
Semin Nephrol ; 40(2): 138-147, 2020 03.
Article in English | MEDLINE | ID: mdl-32303277

ABSTRACT

Histone deacetylases (HDACs) are part of the epigenetic machinery that regulates transcriptional processes. The current paradigm is that HDACs silence gene expression via regulation of histone protein lysine deacetylation, or by forming corepressor complexes with transcription factors. However, HDACs are more than just nuclear proteins, and they can interact and deacetylate a growing number of nonhistone proteins to regulate cellular function. Cancer-field studies have shown that deranged HDAC activity results in uncontrolled proliferation, inflammation, and fibrosis; all pathologies that also may occur in kidney disease. Over the past decade, studies have emerged suggesting that HDAC inhibitors may prevent and potentially treat various models of acute kidney injury. This review focuses on the physiology of kidney HDACs and highlights the recent advances using HDAC inhibitors to potentially treat kidney disease patients.


Subject(s)
Acute Kidney Injury/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Kidney/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Animals , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Epigenesis, Genetic , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Humans , Kidney/drug effects , Reperfusion Injury/complications , Sepsis/complications , Ureteral Obstruction/complications
18.
Am J Physiol Cell Physiol ; 318(6): C1144-C1153, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32267715

ABSTRACT

The skin is essential for terrestrial life. It is responsible for regulating water permeability and functions as a mechanical barrier that protects against environmental insults such as microbial infection, ultraviolet light, injury, and heat and cold, which could damage the cells of the body and compromise survival of the organism. This barrier is provided by the outer layer, the epidermis, which is composed predominantly of keratinocytes; keratinocytes undergo a program of differentiation to form the stratum corneum comprising the cornified squame "bricks" and lipid "mortar." Dysregulation of this differentiation program can result in skin diseases, including psoriasis and nonmelanoma skin cancers, among others. Accumulating evidence in the literature indicates that the water-, glycerol-, and hydrogen peroxide-transporting channel aquaporin-3 (AQP3) plays a key role in various processes involved in keratinocyte function, and abnormalities in this channel have been observed in several human skin diseases. Here, we discuss the data linking AQP3 to keratinocyte proliferation, migration, differentiation, and survival as well as its role in skin properties and functions like hydration, water retention, wound healing, and barrier repair. We also discuss the mechanisms regulating AQP3 levels, localization, and function and the anomalies in AQP3 that are associated with various skin diseases.


Subject(s)
Aquaporin 3/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Psoriasis/metabolism , Water/metabolism , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Epidermis/pathology , Humans , Keratinocytes/pathology , Organism Hydration Status , Permeability , Psoriasis/pathology , Signal Transduction , Wound Healing
19.
Can J Physiol Pharmacol ; 98(9): 604-610, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32083942

ABSTRACT

High salt intake (HS) is associated with obesity and insulin resistance. ET-1, a peptide released in response to HS, inhibits the actions of insulin on cultured adipocytes through ET-1 type B (ETB) receptors; however, the in vivo implications of ETB receptor activation on lipid metabolism and insulin resistance is unknown. We hypothesized that activation of ETB receptors in response to HS intake promotes dyslipidemia and insulin resistance. In normal salt (NS) fed rats, no significant difference in body mass or epididymal fat mass was observed between control and ETB deficient rats. After 2 weeks of HS, ETB-deficient rats had significantly lower body mass and epididymal fat mass compared to controls. Nonfasting plasma glucose was not different between genotypes; however, plasma insulin concentration was significantly lower in ETB-deficient rats compared to controls, suggesting improved insulin sensitivity. In addition, ETB-deficient rats had higher circulating free fatty acids in both NS and HS groups, with no difference in plasma triglycerides between genotypes. In a separate experiment, ETB-deficient rats had significantly lower fasting blood glucose and improved glucose and insulin tolerance compared to controls. These data suggest that ET-1 promotes adipose deposition and insulin resistance via the ETB receptor.


Subject(s)
Dyslipidemias/metabolism , Endothelin-1/metabolism , Insulin Resistance , Insulin/metabolism , Receptor, Endothelin B/deficiency , Adipose Tissue/metabolism , Adiposity , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Body Weight , Disease Models, Animal , Dyslipidemias/blood , Dyslipidemias/etiology , Fatty Acids, Nonesterified/blood , Humans , Insulin/blood , Male , Mutation , Rats , Rats, Transgenic , Receptor, Endothelin B/genetics , Sodium Chloride, Dietary/adverse effects
20.
Am J Physiol Renal Physiol ; 317(3): F547-F559, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31241990

ABSTRACT

The collecting duct (CD) concentrates the urine, thereby maintaining body water volume and plasma osmolality within a normal range. The endocrine hormone arginine vasopressin acts in the CD to increase water permeability via the vasopressin 2 receptor (V2R)-aquaporin (AQP) axis. Recent studies have suggested that autocrine factors may also contribute to the regulation of CD water permeability. Nitric oxide is produced predominantly by nitric oxide synthase 1 (NOS1) in the CD and acts as a diuretic during salt loading. The present study sought to determine whether CD NOS1 regulates diuresis during changes in hydration status. Male and female control and CD NOS1 knockout (CDNOS1KO) mice were hydrated (5% sucrose water), water deprived, or acutely challenged with the V2R agonist desmopressin. In male mice, water deprivation resulted in decreased urine flow and increased plasma osmolality, copeptin concentration, and kidney AQP2 abundance independent of CD NOS1. In female control mice, water deprivation reduced urine flow, increased plasma osmolality and copeptin, but did not significantly change total AQP2; however, there was increased basolateral AQP3 localization. Surprisingly, female CDNOS1KO mice while on the sucrose water presented with symptoms of dehydration. Fibroblast growth factor 21, an endocrine regulator of sweetness preference, was significantly higher in female CDNOS1KO mice, suggesting that this was reducing their drive to drink the sucrose water. With acute desmopressin challenge, female CDNOS1KO mice failed to appropriately concentrate their urine, resulting in higher plasma osmolality than controls. In conclusion, CD NOS1 plays only a minor role in urine-concentrating mechanisms.


Subject(s)
Dehydration/enzymology , Diuresis , Kidney Concentrating Ability , Kidney Tubules, Collecting/enzymology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Animals , Antidiuretic Agents/pharmacology , Aquaporin 2/genetics , Aquaporin 2/metabolism , Aquaporin 3/genetics , Aquaporin 3/metabolism , Deamino Arginine Vasopressin/pharmacology , Dehydration/physiopathology , Disease Models, Animal , Diuresis/drug effects , Female , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Kidney Concentrating Ability/drug effects , Kidney Tubules, Collecting/drug effects , Male , Mice, Knockout , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/genetics , Organism Hydration Status , Osmolar Concentration , Sex Factors , Signal Transduction , Urodynamics , Water Deprivation
SELECTION OF CITATIONS
SEARCH DETAIL
...