Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
Kidney360 ; 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976566

RÉSUMÉ

BACKGROUND: With the growing use of automated peritoneal dialysis (APD), it is important to improve our knowledge of the clinical patterns and physiology of APD treatment sessions. The ultrafiltration (UF) achieved during each cycle of an APD treatment is assumed to be relatively linear if the delivered prescription is the same. We set out to determine if that is indeed the case. METHODS: Single-center, cross-sectional study of prevalent PD patients. All adult APD patients (> 18 years of age), who had been on PD for >3 months, and >3 months on APD were included. Continuous ambulatory PD patients or those with peritonitis within 3 months of the consent date were excluded. Individual treatment data from 7 consecutive APD treatment sessions with consistent dialysate composition for each cycler exchange were collected for each subject. RESULTS: Thirty-nine subjects met the inclusion criteria and were enrolled. The probability of yielding a positive UF was 48.9% for cycle 1, rising to 90.5% by cycle 6. Adjusting for average dextrose concentration, dwell time, fill volume, solute transfer rate, and number of cycles, we observed that cycles 2 through 6 achieved progressively higher UF volumes than cycle 1 (p < 0.001). The first and last cycles demonstrated significantly different cycle UF volumes compared to a middle cycle (-230 ml and 277 ml, respectively, p < 0.001). CONCLUSIONS: We observed a consistent increase in UF volumes achieved per cycle over the course of an APD treatment session with numerous clinical and physiologic implications. This provides the foundation for future studies investigating peritoneal inter-cycle variations and membrane physiology.

2.
Nat Commun ; 15(1): 5144, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886379

RÉSUMÉ

The renal epithelium is sensitive to changes in blood potassium (K+). We identify the basolateral K+ channel, Kir4.2, as a mediator of the proximal tubule response to K+ deficiency. Mice lacking Kir4.2 have a compensated baseline phenotype whereby they increase their distal transport burden to maintain homeostasis. Upon dietary K+ depletion, knockout animals decompensate as evidenced by increased urinary K+ excretion and development of a proximal renal tubular acidosis. Potassium wasting is not proximal in origin but is caused by higher ENaC activity and depends upon increased distal sodium delivery. Three-dimensional imaging reveals Kir4.2 knockouts fail to undergo proximal tubule expansion, while the distal convoluted tubule response is exaggerated. AKT signaling mediates the dietary K+ response, which is blunted in Kir4.2 knockouts. Lastly, we demonstrate in isolated tubules that AKT phosphorylation in response to low K+ depends upon mTORC2 activation by secondary changes in Cl- transport. Data support a proximal role for cell Cl- which, as it does along the distal nephron, responds to K+ changes to activate kinase signaling.


Sujet(s)
Tubules contournés proximaux , Complexe-2 cible mécanistique de la rapamycine , Souris knockout , Canaux potassiques rectifiants entrants , Potassium , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Canaux potassiques rectifiants entrants/métabolisme , Canaux potassiques rectifiants entrants/génétique , Sérine-thréonine kinases TOR/métabolisme , Potassium/métabolisme , Tubules contournés proximaux/métabolisme , Souris , Complexe-2 cible mécanistique de la rapamycine/métabolisme , Complexe-2 cible mécanistique de la rapamycine/génétique , Phosphorylation , Mâle , Chlorures/métabolisme , Souris de lignée C57BL
3.
Curr Opin Nephrol Hypertens ; 33(5): 512-517, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38934092

RÉSUMÉ

PURPOSE OF REVIEW: Highlight the mechanisms through which vasopressin and hypertonic stress regulate protein metabolism. RECENT FINDINGS: Mammals have an 'aestivation-like' response in which hypertonic stress increases muscle catabolism and urea productionVasopressin can directly regulate ureagenesis in the liver and the kidneyIn humans chronic hypertonic stress is associated with premature aging, diabetes, cardiovascular disease, and premature mortality. SUMMARY: There is an evolutionarily conserved 'aestivation-like' response in humans in which hypertonic stress results in activation of the vasopressin system, muscle catabolism, and ureagenesis in order to promote water conservation.


Sujet(s)
Vasopressines , Humains , Animaux , Vasopressines/métabolisme , Urée/métabolisme , Rein/métabolisme , Protéines/métabolisme , Foie/métabolisme
4.
Microorganisms ; 12(5)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38792682

RÉSUMÉ

Emerging data support associations between the depletion of the healthy gut microbiome and aging-related physiological decline and disease. In humans, fecal microbiota transplantation (FMT) has been used successfully to restore gut microbiome structure and function and to treat C. difficile infections, but its application to healthy aging has been scarcely investigated. The marmoset is an excellent model for evaluating microbiome-mediated changes with age and interventional treatments due to their relatively shorter lifespan and many social, behavioral, and physiological functions that mimic human aging. Prior work indicates that FMT is safe in marmosets and may successfully mediate gut microbiome function and host health. This narrative review (1) provides an overview of the rationale for FMT to support healthy aging using the marmoset as a translational geroscience model, (2) summarizes the prior use of FMT in marmosets, (3) outlines a protocol synthesized from prior literature for studying FMT in aging marmosets, and (4) describes limitations, knowledge gaps, and future research needs in this field.

5.
Am J Physiol Renal Physiol ; 326(6): F1091-F1100, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38695074

RÉSUMÉ

We have previously shown that kidney collecting ducts make vasopressin. However, the physiological role of collecting duct-derived vasopressin is uncertain. We hypothesized that collecting duct-derived vasopressin is required for the appropriate concentration of urine. We developed a vasopressin conditional knockout (KO) mouse model wherein Cre recombinase expression induces deletion of arginine vasopressin (Avp) exon 1 in the distal nephron. We then used age-matched 8- to 12-wk-old Avp fl/fl;Ksp-Cre(-) [wild type (WT)] and Avp fl/fl;Ksp-Cre(+) mice for all experiments. We collected urine, serum, and kidney lysates at baseline. We then challenged both WT and knockout (KO) mice with 24-h water restriction, water loading, and administration of the vasopressin type 2 receptor agonist desmopressin (1 µg/kg ip) followed by the vasopressin type 2 receptor antagonist OPC-31260 (10 mg/kg ip). We performed immunofluorescence and immunoblot analysis at baseline and confirmed vasopressin KO in the collecting duct. We found that urinary osmolality (UOsm), plasma Na+, K+, Cl-, blood urea nitrogen, and copeptin were similar in WT vs. KO mice at baseline. Immunoblots of the vasopressin-regulated proteins Na+-K+-2Cl- cotransporter, NaCl cotransporter, and water channel aquaporin-2 showed no difference in expression or phosphorylation at baseline. Following 24-h water restriction, WT and KO mice had no differences in UOsm, plasma Na+, K+, Cl-, blood urea nitrogen, or copeptin. In addition, there were no differences in the rate of urinary concentration or dilution as in WT and KO mice UOsm was nearly identical after desmopressin and OPC-31260 administration. We conclude that collecting duct-derived vasopressin is not essential to appropriately concentrate or dilute urine.NEW & NOTEWORTHY Hypothalamic vasopressin is required for appropriate urinary concentration. However, whether collecting duct-derived vasopressin is involved remains unknown. We developed a novel transgenic mouse model to induce tissue-specific deletion of vasopressin and showed that collecting duct-derived vasopressin is not required to concentrate or dilute urine.


Sujet(s)
Desmopressine , Tubules collecteurs rénaux , Souris knockout , Animaux , Tubules collecteurs rénaux/métabolisme , Tubules collecteurs rénaux/effets des médicaments et des substances chimiques , Desmopressine/pharmacologie , Capacité de concentration rénale/effets des médicaments et des substances chimiques , Arginine vasopressine/métabolisme , Mâle , Antagonistes des récepteurs de l'hormone antidiurétique/pharmacologie , Souris , Aquaporine-2/métabolisme , Aquaporine-2/génétique , Antidiurétiques/pharmacologie , Récepteurs à la vasopressine/génétique , Récepteurs à la vasopressine/métabolisme , Souris de lignée C57BL , Privation hydrique , Concentration osmolaire , Sodium/urine , Sodium/métabolisme , Vasopressines/métabolisme , Benzazépines
6.
Sci Adv ; 10(6): eadi7840, 2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38324689

RÉSUMÉ

Prolonged obstruction of the ureter, which leads to injury of the kidney collecting ducts, results in permanent structural damage, while early reversal allows for repair. Cell structure is defined by the actin cytoskeleton, which is dynamically organized by small Rho guanosine triphosphatases (GTPases). In this study, we identified the Rho GTPase, Rac1, as a driver of postobstructive kidney collecting duct repair. After the relief of ureteric obstruction, Rac1 promoted actin cytoskeletal reconstitution, which was required to maintain normal mitotic morphology allowing for successful cell division. Mechanistically, Rac1 restricted excessive actomyosin activity that stabilized the negative mitotic entry kinase Wee1. This mechanism ensured mechanical G2-M checkpoint stability and prevented premature mitotic entry. The repair defects following injury could be rescued by direct myosin inhibition. Thus, Rac1-dependent control of the actin cytoskeleton integrates with the cell cycle to mediate kidney tubular repair by preventing dysmorphic cells from entering cell division.


Sujet(s)
Tubules collecteurs rénaux , Tubules collecteurs rénaux/métabolisme , Protéine G rac1/métabolisme , Cytosquelette/métabolisme , Actines/métabolisme , Cytosquelette d'actine/métabolisme
7.
Am J Primatol ; 86(4): e23589, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38143428

RÉSUMÉ

Researchers and veterinarians often use hematology and clinical chemistry to evaluate animal health. These biomarkers are relatively easy to obtain, and understanding how they change across healthy aging is critical to clinical care and diagnostics for these animals. We aimed to evaluate how clinical biomarkers from a chemistry profile and complete blood count (CBC) change with age in common marmosets (Callithrix jacchus). We assessed blood samples collected during routine physical exams at the Southwest National Primate Research Center and the University of Texas Health San Antonio marmoset colonies from November 2020-November 2021. We found that chemistry and CBC profiles varied based on facility, sex, and age. Significant changes in albumin, phosphorus/creatinine ratio, albumin/globulin ratio, amylase, creatinine, lymphocyte percent, hematocrit, granulocytes percent, lymphocytes, hemoglobin, red cell distribution width, and platelet distribution width were all reported with advancing age. Aged individuals also demonstrated evidence for changes in liver, kidney, and immune system function compared with younger individuals. Our results suggest there may be regular changes associated with healthy aging in marmosets that are outside of the range typically considered as normal values for healthy young individuals, indicating the potential need for redefined healthy ranges for clinical biomarkers in aged animals. Identifying animals that exhibit values outside of this defined healthy aging reference will allow more accurate diagnostics and treatments for aging colonies.


Sujet(s)
Callithrix , Hématologie , Animaux , Créatinine , Callitrichinae , Albumines , Marqueurs biologiques
9.
Kidney360 ; 4(8): 1174-1180, 2023 08 01.
Article de Anglais | MEDLINE | ID: mdl-37357355

RÉSUMÉ

Vasopressin is a highly conserved peptide hormone that has been traditionally associated with water homeostasis. There is accumulating evidence in both humans and animal models that vasopressin is implicated in the regulation of metabolism. This review focuses on the effects that vasopressin exerts on the regulation of glucose and fatty acids with a particular emphasis on the potential repercussions of metabolic dysregulation in kidney disease.


Sujet(s)
Transduction du signal , Vasopressines , Humains , Animaux , Vasopressines/métabolisme , Glucose/métabolisme , Homéostasie/physiologie
10.
Physiol Rep ; 11(8): e15671, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-37078378

RÉSUMÉ

High sodium and low potassium intake have both been linked to poor cardiovascular health outcomes and increased mortality rates. A combination of the two is thought to be particularly detrimental. While mechanisms are multiple, the kidney is an important target of harmful effects and low potassium influences on both proximal and distal nephron segments are especially potent. We recently reported that a combined high sodium/low potassium diet causes kidney injury and that low potassium in isolation can have similar effects. However, how sodium intake alters this process is not well-understood. Here we tested the hypothesis that a high sodium intake amplifies effects of low dietary potassium on kidney injury. We observed adding high sodium to low potassium caused an expected increase in blood pressure, but did not worsen markers of kidney injury, inflammation, and fibrosis. It also did not increase abundance or phosphorylation of the sodium chloride cotransporter or its regulatory kinases, SPAK and OxSR1, known renal targets of low potassium. Findings support the claim that dietary potassium deficiency, and not high sodium, is a dominant factor affecting kidney injury in animal models of high sodium/low potassium intake. This suggests further investigation is required to identify optimal ranges of sodium and potassium intake in both healthy populations and in those with kidney disease.


Sujet(s)
Maladies du rein , Sodium alimentaire , Animaux , Rein , Sodium , Potassium , Sodium alimentaire/effets indésirables
11.
Am J Physiol Renal Physiol ; 324(6): F521-F531, 2023 06 01.
Article de Anglais | MEDLINE | ID: mdl-36995926

RÉSUMÉ

The objective of this study was to understand the response of mice lacking insulin-regulated aminopeptidase (IRAP) to an acute water load. For mammals to respond appropriately to acute water loading, vasopressin activity needs to decrease. IRAP degrades vasopressin in vivo. Therefore, we hypothesized that mice lacking IRAP have an impaired ability to degrade vasopressin and, thus, have persistent urinary concentration. Age-matched 8- to 12-wk-old IRAP wild-type (WT) and knockout (KO) male mice were used for all experiments. Blood electrolytes and urine osmolality were measured before and 1 h after water load (∼2 mL sterile water via intraperitoneal injection). Urine was collected from IRAP WT and KO mice for urine osmolality measurements at baseline and after 1 h administration of the vasopressin type 2 receptor antagonist OPC-31260 (10 mg/kg ip). Immunofluorescence and immunoblot analysis were performed on kidneys at baseline and after 1 h acute water load. IRAP was expressed in the glomerulus, thick ascending loop of Henle, distal tubule, connecting duct, and collecting duct. IRAP KO mice had elevated urine osmolality compared with WT mice due to higher membrane expression of aquaporin 2 (AQP2), which was restored to that of controls after administration of OPC-31260. IRAP KO mice developed hyponatremia after an acute water load because they were unable to increase free water excretion due to increased surface expression of AQP2. In conclusion, IRAP is required to increase water excretion in response to an acute water load due to persistent vasopressin stimulation of AQP2.NEW & NOTEWORTHY Insulin-regulated aminopeptidase (IRAP) degrades vasopressin, but its role in urinary concentration and dilution is unknown. Here, we show that IRAP-deficient mice have a high urinary osmolality at baseline and are unable to excrete free water in response to water loading. These results reveal a novel regulatory role for IRAP in urine concentration and dilution.


Sujet(s)
Aquaporine-2 , Insuline , Animaux , Mâle , Souris , Aminopeptidases/génétique , Aminopeptidases/métabolisme , Aquaporine-2/génétique , Aquaporine-2/métabolisme , Insuline/métabolisme , Mammifères/métabolisme , Pression osmotique , Vasopressines/pharmacologie , Vasopressines/métabolisme , Eau/métabolisme
12.
Cell Rep ; 41(12): 111840, 2022 12 20.
Article de Anglais | MEDLINE | ID: mdl-36543132

RÉSUMÉ

Inadequate potassium (K+) consumption correlates with increased mortality and poor cardiovascular outcomes. Potassium effects on blood pressure have been described previously; however, whether or not low K+ independently affects kidney disease progression remains unclear. Here, we demonstrate that dietary K+ deficiency causes direct kidney injury. Effects depend on reduced blood K+ and are kidney specific. In response to reduced K+, the channel Kir4.2 mediates altered proximal tubule (PT) basolateral K+ flux, causing intracellular acidosis and activation of the enzyme glutaminase and the ammoniagenesis pathway. Deletion of either Kir4.2 or glutaminase protects from low-K+ injury. Reduced K+ also mediates injury and fibrosis in a model of aldosteronism. These results demonstrate that the PT epithelium, like the distal nephron, is K+ sensitive, with reduced blood K+ causing direct PT injury. Kir4.2 and glutaminase are essential mediators of this injury process, and we identify their potential for future targeting in the treatment of chronic kidney disease.


Sujet(s)
Canaux potassiques rectifiants entrants , Potassium , Potassium/métabolisme , Glutaminase/génétique , Glutaminase/métabolisme , Canaux potassiques rectifiants entrants/métabolisme , Rein/métabolisme , Néphrons/métabolisme
13.
JCI Insight ; 7(24)2022 12 22.
Article de Anglais | MEDLINE | ID: mdl-36326835

RÉSUMÉ

Vasopressin has traditionally been thought to be produced by the neurohypophyseal system and then released into the circulation where it regulates water homeostasis. The questions of whether vasopressin could be produced outside of the brain and if the kidney could be a source of vasopressin are raised by the syndrome of inappropriate antidiuretic hormone secretion (vasopressin). We found that mouse and human kidneys expressed vasopressin mRNA. Using an antibody that detects preprovasopressin, we found that immunoreactive preprovasopressin protein was found in mouse and human kidneys. Moreover, we found that murine collecting duct cells made biologically active vasopressin, which increased in response to NaCl-mediated hypertonicity, and that water restriction increased the abundance of kidney-derived vasopressin mRNA and protein expression in mouse kidneys. Thus, we provide evidence of biologically active production of kidney-derived vasopressin in kidney tubular epithelial cells.


Sujet(s)
Tubules collecteurs rénaux , Souris , Humains , Animaux , Tubules collecteurs rénaux/métabolisme , Chlorure de sodium/pharmacologie , Chlorure de sodium/métabolisme , Vasopressines/métabolisme , Eau/métabolisme , ARN messager/métabolisme
15.
JCI Insight ; 7(4)2022 02 22.
Article de Anglais | MEDLINE | ID: mdl-35025763

RÉSUMÉ

Aristolochic acid (AA) is the causative nephrotoxic alkaloid in AA nephropathy, which results in a tubulointerstitial fibrosis. AA causes direct proximal tubule damage as well as an influx of macrophages, although the role of macrophages in pathogenesis is poorly understood. Here, we demonstrate that AA directly stimulates migration, inflammation, and ROS production in macrophages ex vivo. Cells lacking interferon regulatory factor 4 (IRF4), a known regulator of macrophage migration and phenotype, had a reduced migratory response, though effects on ROS production and inflammation were preserved or increased relative to WT cells. Macrophage-specific IRF4-knockout mice were protected from both acute and chronic kidney effects of AA administration based on functional and histological analysis. Renal macrophages from kidneys of AA-treated macrophage-specific IRF4-knockout mice demonstrated increased apoptosis and ROS production compared with WT controls, indicating that AA directly polarizes macrophages to a promigratory and proinflammatory phenotype. However, knockout mice had reduced renal macrophage abundance following AA administration. While macrophages lacking IRF4 can adopt a proinflammatory phenotype upon AA exposure, their inability to migrate to the kidney and increased rates of apoptosis upon infiltration provide protection from AA in vivo. These results provide evidence of direct AA effects on macrophages in AA nephropathy and add to the growing body of evidence that supports a key role of IRF4 in modulating macrophage function in kidney injury.


Sujet(s)
Apoptose , ADN/génétique , Facteurs de régulation d'interféron/génétique , Tubules contournés proximaux/métabolisme , Macrophages/métabolisme , Mutation , Insuffisance rénale chronique/génétique , Animaux , Acides aristolochiques/toxicité , Cellules cultivées , Analyse de mutations d'ADN , Modèles animaux de maladie humaine , Fibrose/génétique , Fibrose/métabolisme , Fibrose/anatomopathologie , Délétion de gène , Facteurs de régulation d'interféron/métabolisme , Tubules contournés proximaux/anatomopathologie , Macrophages/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Insuffisance rénale chronique/induit chimiquement , Insuffisance rénale chronique/anatomopathologie
16.
Kidney Int ; 100(4): 894-905, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34111501

RÉSUMÉ

Acute kidney injury is a common complication in patients hospitalized with SARSCoV-2 (COVID-19), with prior studies implicating multiple potential mechanisms of injury. Although COVID-19 is often compared to other respiratory viral illnesses, few formal comparisons of these viruses on kidney health exist. In this retrospective cohort study, we compared the incidence, features, and outcomes of acute kidney injury among Veterans hospitalized with COVID-19 or influenza and adjusted for baseline conditions using weighted comparisons. A total of 3402 hospitalizations for COVID-19 and 3680 hospitalizations for influenza admitted between October 1, 2019 and May 31, 2020 across 127 Veterans Administration hospitals nationally were studied using the electronic medical record. Acute kidney injury occurred more frequently among those with COVID-19 compared to those with influenza (40.9% versus 29.4%, weighted analysis) and was more severe. Patients with COVID-19 were more likely to require mechanical ventilation and vasopressors and experienced higher mortality. Proteinuria and hematuria were frequent in both groups but more common in COVID-19. Recovery of kidney function was less common in patients with COVID-19 and acute kidney injury but was similar among survivors. Thus, findings from this study confirm that acute kidney injury is more common and severe among patients hospitalized with COVID-19 compared to influenza, a finding that may be driven largely by illness severity. Hence, the combined impact of these two illnesses on kidney health may be significant and have important implications for resource allocation.


Sujet(s)
Atteinte rénale aigüe , COVID-19 , Grippe humaine , Anciens combattants , Atteinte rénale aigüe/diagnostic , Atteinte rénale aigüe/épidémiologie , Atteinte rénale aigüe/thérapie , Mortalité hospitalière , Humains , Incidence , Grippe humaine/épidémiologie , Études rétrospectives , SARS-CoV-2 , États-Unis/épidémiologie
17.
Am J Physiol Renal Physiol ; 320(4): F569-F577, 2021 04 01.
Article de Anglais | MEDLINE | ID: mdl-33522414

RÉSUMÉ

Ischemic heart disease is the leading cause of death worldwide and is frequently comorbid with chronic kidney disease. Physiological communication is known to occur between the heart and the kidney. Although primary dysfunction in either organ can induce dysfunction in the other, a clinical entity known as cardiorenal syndrome, mechanistic details are lacking. Here, we used a model of experimental myocardial infarction (MI) to test effects of chronic cardiac ischemia on acute and chronic kidney injury. Surprisingly, chronic cardiac damage protected animals from subsequent acute ischemic renal injury, an effect that was accompanied by evidence of chronic kidney hypoxia. The protection observed post-MI was similar to protection observed in a separate group of healthy animals housed in ambient hypoxic conditions prior to kidney injury, suggesting a common mechanism. There was evidence that chronic cardiac injury activates renal hypoxia-sensing pathways. Increased renal abundance of several glycolytic enzymes following MI suggested that a shift toward glycolysis may confer renal ischemic preconditioning. In contrast, effects on chronic renal injury followed a different pattern, with post-MI animals displaying worsened chronic renal injury and fibrosis. These data show that although chronic cardiac injury following MI protected against acute kidney injury via activation of hypoxia-sensing pathways, it worsened chronic kidney injury. The results further our understanding of cardiorenal signaling mechanisms and have implications for the treatment of heart failure patients with associated renal disease.NEW & NOTEWORTHY Experimental myocardial infarction (MI) protects from subsequent ischemic acute kidney injury but worsens chronic kidney injury. Observed protection from ischemic acute kidney injury after MI was accompanied by chronic kidney hypoxia and increased renal abundance of hypoxia-inducible transcripts. These data support the idea that MI confers protection from renal ischemic injury via chronic renal hypoxia and activation of downstream hypoxia-inducible signaling pathways.


Sujet(s)
Atteinte rénale aigüe/métabolisme , Syndrome cardiorénal/complications , Hypoxie/métabolisme , Préconditionnement ischémique , Infarctus du myocarde/complications , Atteinte rénale aigüe/complications , Atteinte rénale aigüe/anatomopathologie , Animaux , Syndrome cardiorénal/physiopathologie , Coeur/physiopathologie , Défaillance cardiaque/métabolisme , Rein/métabolisme , Infarctus du myocarde/métabolisme , Myocarde/anatomopathologie , Insuffisance rénale chronique/complications , Insuffisance rénale chronique/métabolisme
18.
Kidney360 ; 2(9): 1434-1440, 2021 09 30.
Article de Anglais | MEDLINE | ID: mdl-35373107

RÉSUMÉ

Background: Insulin resistance is associated with cardiovascular disease risk and worsened kidney function. Patients with CKD have higher levels of insulin resistance. Elevated levels of copeptin (a surrogate for vasopressin levels) have been associated with an increased incidence and progression of CKD, and with incident diabetes mellitus. The purpose of our study was to examine the relationship between insulin resistance, copeptin, and CKD. Methods: We performed a cross-sectional study to investigate if insulin resistance was associated with higher copeptin levels in nondiabetic patients with stage 3-4 CKD versus controls. We measured plasma copeptin levels and used data from 52 patients with stage 3-4 CKD and 85 controls (eGFR ≥60 ml/min per 1.73 m2) enrolled in the Insulin Resistance in Chronic Kidney Disease (IRCKD) study. We then used a multivariable linear-regression model to assess the independent relationship between peripheral or hepatic insulin resistance and copeptin across levels of eGFR. Results: We found that in patients with CKD (eGFR of 30-60 ml/min per 1.73 m2), but not in controls, peripheral insulin resistance was significantly correlated with higher levels of log copeptin (r=-0.21, P=0.04). In patients with CKD, when adjusted for age, sex, BMI, serum osmolality, log IL6, and log leptin/adiponectin ratio, each 1 SD decrease in insulin sensitivity was associated with a 39% increase in serum copeptin levels. The relationship between hepatic insulin resistance, copeptin, and eGFR is similar between controls and patients with reduced eGFR. Conclusion: Peripheral insulin resistance is associated with elevated copeptin levels in nondiabetic patients with stage 3-4 CKD. Further research into how the interaction between peripheral insulin resistance and elevated vasopressin affects CKD progression could be of interest.


Sujet(s)
Insulinorésistance , Insuffisance rénale chronique , Études transversales , Glycopeptides , Humains , Insuline
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE