1
|
Coppola S, Chiumello D, Adnan A, Pozzi T, Forni LG, Gattinoni L. Diuretics in critically ill patients: a narrative review of their mechanisms and applications. Br J Anaesth 2025; 134:1638-1647. [PMID: 40221314 DOI: 10.1016/j.bja.2025.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/30/2025] [Accepted: 02/27/2025] [Indexed: 04/14/2025] Open
Abstract
Diuretics remain the cornerstone therapy of critically ill patients with volume overload as a result of cardiac failure, acute kidney injury or aggressive fluid resuscitation. This review summarises the principles of applied renal physiology, describing the mechanisms of action, the clinical applications, and the adverse effects of commonly used diuretics during critical illness. Loop diuretics, and in particular furosemide, remain the most popular, despite evidence of any effect on mortality or, indeed, on the need for renal replacement therapy. The efficacy of loop diuretics after administration depends on three factors. Firstly, the tubular concentration of the diuretic: continuous infusion of furosemide seems to provide a higher and more stable tubular concentration of furosemide with respect to bolus injection. Secondly, the interaction with albumin both in the plasma and in the renal tubule: despite a strong physiological rationale supporting this approach, albumin supplementation in hypoalbuminaemic patients does not seem to result in a higher diuretic efficacy. Thirdly, diuretic resistance, which can be addressed by optimising loop diuretic dose and by using combination therapy with other agents, including thiazides or thiazide-like diuretics or carbonic anhydrase inhibitors. These drugs constitute a useful adjunct to overcome loop diuretic resistance. Other agents such as distal potassium-sparing diuretics and osmotic diuretics can also be considered. The latter have been used successfully in hypokalaemia, rhabdomyolysis-associated acute kidney injury or to prevent ischaemia-reperfusion injury in kidney transplantation. Finally, this review provides the basic concepts of the interplay between acid-base equilibrium and diuretic therapy.
Collapse
Affiliation(s)
- Silvia Coppola
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Milan, Italy
| | - Davide Chiumello
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy; Coordinated Research Center on Respiratory Failure, University of Milan, Milan, Italy.
| | - Afiqah Adnan
- Royal Surrey Hospital NHS Foundation Trust, Guildford, UK
| | - Tommaso Pozzi
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Milan, Italy; Department of Health Sciences, University of Milan, Milan, Italy
| | - Lui G Forni
- Royal Surrey Hospital NHS Foundation Trust, Guildford, UK; School of Medicine, University of Surrey, Kate Granger Building, Guildford, UK
| | - Luciano Gattinoni
- Department of Anaesthesiology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
2
|
Keller C, Santos RR, van Megen WH, Loffing J. Characterization of ROMK cellular heterogeneity along the mouse kidney thick ascending limb. Pflugers Arch 2025; 477:841-856. [PMID: 40358700 DOI: 10.1007/s00424-025-03086-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
The renal thick ascending limb (TAL) plays a key role in water and ion homeostasis. Apical potassium secretion via the renal outer medullary potassium channel (ROMK) is essential for transepithelial sodium reabsorption via the furosemide-sensitive Na-K-2Cl-cotransporter and creates the electrochemical gradient for paracellular ion transport through Claudin tight junction proteins. Interestingly, the TAL exhibits transcriptomic heterogeneity and variable apical ROMK abundance. Single-cell RNA sequencing suggests that the cortical TAL consists of at least three distinct cell types, but whether ROMK distribution aligns with these types remains unclear. We analyzed perfusion-fixed mouse kidneys using RNAscope in situ hybridization (ISH), iterative indirect immunofluorescence imaging (4i multiplexing), and machine learning. ROMK mRNA expression was seen in all TAL cells. In contrast, apical ROMK protein abundance was found on almost all macula densa (MD) cells but was heterogeneous along the rest of the TAL. In the remaining TAL, only about 60% of the TAL cells had strong apical ROMK staining, while 40% lacked apical ROMK but showed weak perinuclear signals. ISH revealed that apical ROMK-positive cells express Ptger3 mRNA, whereas apical ROMK-negative cells express Foxq1 mRNA. Multiplexing analysis showed that ROMK-positive cells form Claudin-10b-positive tight junctions, while ROMK-negative cells form Claudin-16/19-positive junctions and express basolateral Kir4.1. Despite universal ROMK mRNA expression, apical ROMK distribution aligns with molecularly distinct TAL cell types. This unique ROMK expression pattern suggests functional heterogeneity for ROMK along the TAL.
Collapse
Affiliation(s)
- Christian Keller
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH- 8057, Zurich, Switzerland
- PhD Program Biomedicine, University of Zurich, Zurich, Switzerland
| | - Rui Ramos Santos
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH- 8057, Zurich, Switzerland
- Ophthalmology Clinic, City Hospital Zurich, Zurich, Switzerland
- Spross Research Institute, Zurich, Switzerland
| | - Wouter H van Megen
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH- 8057, Zurich, Switzerland
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH- 8057, Zurich, Switzerland.
| |
Collapse
|
3
|
Gallegos FR, Delahunty MP, Hu J, Yerigeri SB, Dev V, Bhatt G, Raina R. Decoding Monogenic Hypertension: A Review of Rare Hypertension Disorders. Am J Hypertens 2025; 38:333-351. [PMID: 39803890 DOI: 10.1093/ajh/hpaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/23/2024] [Accepted: 01/02/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Hypertension is a growing concern worldwide, with increasing prevalence rates in both children and adults. Most cases of hypertension are multifactorial, with various genetic, environmental, socioeconomic, and lifestyle influences. However, monogenic hypertension, a blanket term for a group of rare hypertensive disorders, is caused by single-gene mutations that are typically inherited in an autosomal dominant fashion, and ultimately disrupt normal blood pressure regulation in the kidney or adrenal gland. Being able to recognize and understand the pathophysiology of these rare disorders is critical for properly diagnosing hypertension, particularly in children and young adults, as treating each form of monogenic hypertension requires specific and targeted treatment approaches. METHODS A scoping literature review was conducted on the available knowledge regarding each of the disorders currently categorized as forms of monogenic hypertension. RESULTS This narrative review serves to highlight the epidemiology, pathophysiology, clinical presentation, recent case reports, and most current methods of evaluation and treatment for familial hyperaldosteronism types 1-4, Gordon syndrome. Liddle syndrome, syndrome of apparent mineralocorticoid excess, congenital adrenal hyperplasia, Geller syndrome, hereditary syndromes related to pheochromocytomas and paragangliomas, and brachydactyly type E. CONCLUSIONS Recent and future advances in genetic analysis techniques will further enhance the diagnosis and early management of these disorders, preventing the consequences of uncontrolled hypertension.
Collapse
Affiliation(s)
- Flora R Gallegos
- Department of Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Meaghan P Delahunty
- Department of Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Jieji Hu
- Department of Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Shivani B Yerigeri
- Department of Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Vishnu Dev
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Girish Bhatt
- Division of Pediatric Nephrology, Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), Bhopal, India
| | - Rupesh Raina
- Department of Medicine, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio, USA
- Department of Pediatric Nephrology, Akron Children's Hospital, Akron, Ohio, USA
- Department of Internal Medicine, Summa Health System, Akron, Ohio, USA
| |
Collapse
|
4
|
Babu A, Hulse WN, Harer MW, Drake KA, Kling PJ. Developmental origins of disease - Effects of iron deficiency in the rat developing kidney and beyond. Pediatr Nephrol 2025:10.1007/s00467-025-06762-w. [PMID: 40220077 DOI: 10.1007/s00467-025-06762-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/14/2025]
Abstract
Iron is an essential cofactor in metabolic and developmental processes. Iron deficiency (ID) is the most common micronutrient deficiency in pregnancy, especially impacting medically underserved populations worldwide. Iron deficiency (ID) in pregnancy predisposes neonates to poor iron status, i.e., congenital ID and associated adverse effects. The role of congenital ID on human kidney development is unstudied, but impaired fetal kidney development is possible. Both vascular and global nutrient restriction rat models report impaired fetal kidney development, as well as induce hypertension, supporting the developmental origins of health and disease (DOHaD) hypothesis. This review compiles findings from 17 published studies in rats examining congenital or early postnatal ID, showing the same. The review compares histological and physiological findings in both congenital and postnatal ID, placing these in the context of recent knowledge describing molecular mechanistic pathways regulating nephrogenesis. Findings in rat early-life ID include lower kidney iron levels, lower glomerular generations and estimated glomerular numbers, larger maculae densa size, interstitial fibrosis, and prolonging active glomerulogenesis past normal temporal cessation. Additionally, several physiological studies in rat congenital ID promote altered renin-angiotensin signaling and hypertension with maturation, especially in males. Key findings of morphological kidney maldevelopment, altered renin-angiotensin signaling, and hypertension in early-life ID underscore the urgent need for future mechanistic data in animals such as rats. The long-term goal would be to leverage understanding from these data into either preventative or early therapeutic strategies in children.
Collapse
Affiliation(s)
- Anthony Babu
- Division of Neonatology, University of Wisconsin-Madison Department of Pediatrics, Madison, WI, USA
| | - Whitley N Hulse
- Division of Neonatology, University of Wisconsin-Madison Department of Pediatrics, Madison, WI, USA
- Unitypoint Health Meriter, 202 S. Park St., Madison, WI, 53715, USA
| | - Matthew W Harer
- Division of Neonatology, University of Wisconsin-Madison Department of Pediatrics, Madison, WI, USA
- Unitypoint Health Meriter, 202 S. Park St., Madison, WI, 53715, USA
| | - Keri A Drake
- Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Pamela J Kling
- Division of Neonatology, University of Wisconsin-Madison Department of Pediatrics, Madison, WI, USA.
- Unitypoint Health Meriter, 202 S. Park St., Madison, WI, 53715, USA.
| |
Collapse
|
5
|
Gross O, Boeckhaus J, Weber LT, Heerspink HJL, Simon JF, Ahmed R, Gerst C, Duerr U, Walker F, Tostmann R, Helm J, Asendorf T, Friede T. Protocol and rationale for a randomized controlled SGLT2 inhibitor trial in paediatric and young adult populations with chronic kidney disease: DOUBLE PRO-TECT Alport. Nephrol Dial Transplant 2025; 40:679-687. [PMID: 39122650 PMCID: PMC11960741 DOI: 10.1093/ndt/gfae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Clinical trials have demonstrated positive cardiovascular and kidney outcomes of sodium-glucose co-transporter 2 (SGLT2) inhibitors in adult patients with diabetic and other chronic kidney diseases (CKDs). Whether benefits extend to children, teenagers and young adults with early-stage CKD is unknown. For this reason, the DOUBLE PRO-TECT Alport trial (NCT05944016) will study the progression of albuminuria in young patients with Alport syndrome (AS), the most common hereditary CKD, to assess the safety and efficacy of the SGLT2 inhibitor dapagliflozin. Patients living with AS and chronically elevated albuminuria have a high risk of kidney failure before the age of 50 years. METHODS DOUBLE PRO-TECT Alport is a multicentre, randomized, double-blind, placebo-controlled trial. Participants (ages 10-39 years) must have a diagnosis of AS by genetic testing or kidney biopsy, be on a stable (>3 months) maximum tolerated dose of a renin-angiotensin system inhibitor and have a urinary albumin:creatinine ratio (UACR) of >300 mg/g (paediatric) or >500 mg/g (adult).Eligible participants will be randomly assigned at a 2:1 ratio to 48 weeks of treatment with dapaglifozin 10 mg/day or matched placebo. Most participants are expected to be children with a normal estimated glomerular filtration rate (eGFR). In addition to safety, the primary (change in UACR from baseline to week 48) and key secondary (eGFR change from baseline to week 52) efficacy outcomes will be analysed with a mixed model repeated measures approach. Efficacy analyses will be performed primarily in the full analysis set according to the intention-to-treat principle. A sensitivity analysis will be performed using reference-based multiple imputation. CONCLUSION DOUBLE PRO-TECT Alport will assess whether SGLT2 inhibitors can safely reduce the UACR change from baseline as a marker for progression of CKD in young patients living with AS.
Collapse
Affiliation(s)
- Oliver Gross
- Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Jan Boeckhaus
- Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
| | - Lutz T Weber
- Pediatric Nephrology, Children's and Adolescents’ Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - James F Simon
- Department of Kidney Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Rees Ahmed
- Legal Department, University Medical Center Göttingen, Göttingen, Germany
| | - Christoph Gerst
- Legal Department, University Medical Center Göttingen, Göttingen, Germany
| | - Ulrike Duerr
- Nephrology and Rheumatology, University Medical Center Göttingen, Göttingen, Germany
- Clinical Trials Unit, University Medical Center Göttingen, Göttingen, Germany
| | - Florian Walker
- Clinical Trials Unit, University Medical Center Göttingen, Göttingen, Germany
| | - Ralf Tostmann
- Clinical Trials Unit, University Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Helm
- Clinical Trials Unit, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Asendorf
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Tim Friede
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
6
|
Acera-Mateos M, Adiconis X, Li JK, Marchese D, Caratù G, Hon CC, Tiwari P, Kojima M, Vieth B, Murphy MA, Simmons SK, Lefevre T, Claes I, O'Connor CL, Menon R, Otto EA, Ando Y, Vandereyken K, Kretzler M, Bitzer M, Fraenkel E, Voet T, Enard W, Carninci P, Heyn H, Levin JZ, Mereu E. Systematic evaluation of single-cell multimodal data integration for comprehensive human reference atlas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.637075. [PMID: 40093094 PMCID: PMC11908249 DOI: 10.1101/2025.03.06.637075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The integration of multimodal single-cell data enables comprehensive organ reference atlases, yet its impact remains largely unexplored, particularly in complex tissues. We generated a benchmarking dataset for the renal cortex by integrating 3' and 5' scRNA-seq with joint snRNA-seq and snATAC-seq, profiling 119,744 high-quality nuclei/cells from 19 donors. To align cell identities and enable consistent comparisons, we developed the interpretable machine learning tool scOMM (single-cell Omics Multimodal Mapping) and systematically assessed integration strategies. "Horizontal" integration of scRNA and snRNA-seq improved cell-type identification, while "vertical" integration of snRNA-seq and snATAC-seq had an additive effect, enhancing resolution in homogeneous populations and difficult-to-identify states. Global integration was especially effective in identifying adaptive states and rare cell types, including WFDC2-expressing Thick Ascending Limb and Norn cells, previously undetected in kidney atlases. Our work establishes a robust framework for multimodal reference atlas generation, advancing single-cell analysis and extending its applicability to diverse tissues.
Collapse
Affiliation(s)
- Mario Acera-Mateos
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Xian Adiconis
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Ginevra Caratù
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - Chung-Chau Hon
- Laboratory for Regulatory Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Prabha Tiwari
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Miki Kojima
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Beate Vieth
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Planegg, Germany
| | - Michael A Murphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Current affiliation: Osmo; New York, NY 10016, USA
| | - Sean K Simmons
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas Lefevre
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
| | - Irene Claes
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
| | - Christopher L O'Connor
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rajasree Menon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Edgar A Otto
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yoshinari Ando
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Katy Vandereyken
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
| | - Matthias Kretzler
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Markus Bitzer
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Thierry Voet
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Planegg, Germany
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Human Technopole, Milano, Italy
| | - Holger Heyn
- University of Barcelona (UB), Barcelona, Spain
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Joshua Z Levin
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | |
Collapse
|
7
|
Hu T, Li L, Cao Q, Tu W, Huang X, Yuan T. Positive association between serum lactate dehydrogenase levels and blood pressure: evidence from NHANES 2015-2016. Front Cardiovasc Med 2025; 12:1554702. [PMID: 40094026 PMCID: PMC11906999 DOI: 10.3389/fcvm.2025.1554702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Background Serum lactate dehydrogenase (sLDH) is an enzyme implicated in tissue injury and inflammatory responses. Despite its established role in these pathophysiological processes, the association between sLDH and blood pressure remains underexplored. The present findings suggest that sLDH could emerge as a valuable biomarker for blood pressure regulation and may hold significant promise in the management of hypertension. Methods Our investigation utilized data from the National Health and Nutrition Examination Survey (NHANES) 2015-2016, comprising 3,469 participants after excluding those under the age of 20, individuals on antihypertensive therapies, and cases with incomplete data. sLDH levels were categorized into tertiles, while blood pressure measurements were conducted under standardized protocols. To elucidate the relationship between sLDH levels and blood pressure, multivariate regression analyses and smooth curve fitting techniques were employed, adjusting for 17 covariates, including age, sex, and body mass index. Results sLDH corresponds with both systolic blood pressure (SBP) and diastolic blood pressure (DBP). The adjusted smooth curve fitting diagram demonstrates a linear positive connection between sLDH and SBP, with an increment of 0.053 mmHg (95% CI: 0.032, 0.074; p < 0.001) in SBP for every 1 U/L increment in LDH concentrations. The connection between sLDH and DBP is non-linear. sLDH concentrations below 123 U/L have a linear positive connection with DBP, increasing 0.079 mmHg (95% CI: 0.042, 0.115, p < 0.001). When sLDH concentrations exceed 123 U/L, there is not a substantial connection with DBP (P = 0.574). Conclusion Our study demonstrates a linear positive correlation between sLDH and SBP. A non-linear association was observed between sLDH and DBP, with a positive relationship for sLDH levels below 123 U/L. These findings underscore the potential of sLDH as a biomarker for blood pressure regulation.
Collapse
Affiliation(s)
- Tao Hu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Linfeng Li
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qiqiang Cao
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Weiling Tu
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - XianTao Huang
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Tan Yuan
- Department of Cardiology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Guzik M, Zymliński R, Ponikowski P, Biegus J. Urine chloride trajectory and relationship with diuretic response in acute heart failure. ESC Heart Fail 2025; 12:133-141. [PMID: 39438405 PMCID: PMC11769642 DOI: 10.1002/ehf2.15054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/31/2024] [Accepted: 08/21/2024] [Indexed: 10/25/2024] Open
Abstract
AIMS Sodium excretion is a well-defined marker used to assess diuretic response in acute heart failure (AHF). Despite a strong pathophysiological background, the role of urine chloride excretion has not been described and established yet. We aimed to evaluate chloride trajectory during intensive diuretic treatment in AHF patients and examine its potential role in predicting poor diuretic response. METHODS The study was conducted on 50 AHF patients. Participants were included within the first 36 h of hospitalization. They received furosemide dose adjusted for body weight (half in bolus, half in 2 h infusion). Post-diuretic hourly urine collection with biochemical analysis was performed. RESULTS In general, the concentrations of urine chloride (uCl-) and sodium (uNa+) at the baseline samples exhibited a comparable level (71 ± 39 vs. 70 ± 44 mmol/L, respectively; P = 0.99), but across all post-furosemide study timepoints, uCl- remained significantly higher than uNa+ since 1 to 6 h of the study. In this course, both ions (uCl- and uNa+) reached peak values in 2 h (114 ± 28 vs. 97 ± 34 mmol/L, respectively; P < 0.01). The pattern of uCl- dominance over uNa+ concentration was also observed in separate analyses of patients naïve to furosemide and those chronically exposed to furosemide. Regardless of these patterns, naïve to furosemide individuals excreted more ions (both uCl- and uNa+) than chronically exposed patients at all timepoints. Additionally, a strong, linear correlation between uCl- and uNa+ was observed in each post-furosemide timepoint (the strongest in 1 h r = 0.87; P < 0.001). Both interdependent ions concentration was almost parallel when analysed in chronic furosemide users and those naïve to furosemide separately [uCl- = 0.85 * uNa+ + 28.82, P < 0.001, R2 = 0.83 for chronic furosemide users, and uCl- = 0.72 * uNa+ + 41.55, P < 0.001, R2 = 0.65 for naïves to furosemide (linear regression model)]. Moreover, uCl- (with cutoff point: 72 mmol/L) was a satisfactory predictive factor for poor diuretic response (<100 mL/h in 6 h since the beginning of furosemide infusion) [odds ratio (OR) 95% confidence interval (CI): 39.0 (3.8-405.00)]. It presented those properties also after adjusting for urine creatinine [cutoff point: 0.296 mmol/mg-OR (95% CI): 81.0 (8.0-816.0)]. CONCLUSIONS Urine chloride and sodium are highly interrelated during decongestion of AHF patients. The uCl- (cutoff 72 mmol/L) exhibits better prognostic abilities to identify poor diuretic response than uNa+.
Collapse
Affiliation(s)
| | | | | | - Jan Biegus
- Institute of Heart DiseasesWroclawPoland
| |
Collapse
|
9
|
Lin CW, Hung SY, Chen IW. Analysis of the Endocrine Responses to Anti-Diabetes Drugs: An Issue of Elevated Plasma Renin Concentration in Sodium-Glucose Co-Transporter 2 Inhibitor. Int J Gen Med 2025; 18:135-144. [PMID: 39816640 PMCID: PMC11734502 DOI: 10.2147/ijgm.s497664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Purpose Glucose metabolism is associated with several endocrine disorders. Anti-diabetes drugs are crucial in controlling diabetes and its complications; nevertheless, few studies have been carried out involving endocrine function. This study aimed to investigate the association between anti-diabetes drugs and endocrine parameters. Patients and Methods We performed a study of 180 consecutive patients with type 2 diabetes who attended a medical center. Laboratory measurements of metabolic values and endocrine parameters were assessed after a stable treatment regimen of more than 12 weeks. The differences in various endocrine parameters were compared between subjects with or without certain anti-diabetes drugs, with the administrated anti-diabetes drugs being analyzed to find independent risks associated with elevated endocrine parameters. Results After maintaining stable treatment, acceptable glycemic control was noted with an average HbA1c of 7.55% in females and 7.43% in males. Participants taking sulfonylurea (55.8 vs 26.34 ng/L, P=0.043), dipeptidyl peptidase-4 inhibitor (DPP4i) (47.14 vs 32.26 ng/L, P=0.096), or sodium-glucose co-transporter 2 inhibitor (SGLT2i) (64.58 vs 28.11 ng/L, P=0.117) had higher plasma renin concentrations compared to those without this drug but the aldosterone levels did not differ, as well as for other adrenal tests and thyroid function. Under linear regression modeling, SGLT2i was found to be independently associated with a risk of high renin level (beta coefficient: 30.186, 95% confidence interval: 1.71─58.662, P=0.038), whereas sulfonylurea only had borderline associations (B: 21.143, 95% CI: -2.729─45.014, P=0.082). Additionally, renin-angiotensin-aldosterone system (RAAS) blockade (B: 36.728, 95% CI: 12.16─61.295, P=0.004) or diuretics (B: 47.847, 95% CI: 2.039─93.655, P=0.041) was also independently associated with increased renin levels. Conclusion SGLT2i was the only class of anti-diabetes drugs independently associated with elevated renin levels, with results similar to RAAS blockade and diuretics. Although SGLT2i appears to protect reno- and cardio-function, the clinical impact of increased renin warrants further precise study for verification.
Collapse
Affiliation(s)
- Cheng-Wei Lin
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu City, Taiwan
| | - Shih-Yuan Hung
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu City, Taiwan
| | - I-Wen Chen
- Division of Endocrinology and Metabolism, Chang Gung Memorial Hospital at Linkou, Taoyuan City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| |
Collapse
|
10
|
Shankland SJ, Pippin JW, Wessely O. A contemporary view of the macula densa: kidney remodeling. Kidney Int 2025; 107:7-9. [PMID: 39419482 DOI: 10.1016/j.kint.2024.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/12/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Stuart J Shankland
- Division of Nephrology, University of Washington, Seattle, Washington, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA.
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington, USA
| | - Oliver Wessely
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA.
| |
Collapse
|
11
|
Hornung E, Achanta S, Moss A, Schwaber JS, Vadigepalli R. Multi-organ gene expression analysis and network modeling reveal regulatory control cascades during the development of hypertension in female spontaneously hypertensive rat. PLoS One 2024; 19:e0313252. [PMID: 39514592 PMCID: PMC11548744 DOI: 10.1371/journal.pone.0313252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Hypertension is a multifactorial disease with stage-specific gene expression changes occurring in multiple organs over time. The temporal sequence and the extent of gene regulatory network changes occurring across organs during the development of hypertension remain unresolved. In this study, female spontaneously hypertensive (SHR) and normotensive Wistar Kyoto (WKY) rats were used to analyze expression patterns of 96 genes spanning inflammatory, metabolic, sympathetic, fibrotic, and renin-angiotensin (RAS) pathways in five organs, at five time points from the onset to established hypertension. We analyzed this multi-dimensional dataset containing ~15,000 data points and developed a data-driven dynamic network model that accounts for gene regulatory influences within and across visceral organs and multiple brainstem autonomic control regions. We integrated the data from female SHR and WKY with published multiorgan gene expression data from male SHR and WKY. In female SHR, catecholaminergic processes in the adrenal gland showed the earliest gene expression changes prior to inflammation-related gene expression changes in the kidney and liver. Hypertension pathogenesis in male SHR instead manifested early as catecholaminergic gene expression changes in brainstem and kidney, followed by an upregulation of inflammation-related genes in liver. RAS-related gene expression from the kidney-liver-lung axis was downregulated and intra-adrenal RAS was upregulated in female SHR, whereas the opposite pattern of gene regulation was observed in male SHR. We identified disease-specific and sex-specific differences in regulatory interactions within and across organs. The inferred multi-organ network model suggests a diminished influence of central autonomic neural circuits over multi-organ gene expression changes in female SHR. Our results point to the gene regulatory influence of the adrenal gland on spleen in female SHR, as compared to brainstem influence on kidney in male SHR. Our integrated molecular profiling and network modeling identified a stage-specific, sex-dependent, multi-organ cascade of gene regulation during the development of hypertension.
Collapse
Affiliation(s)
- Eden Hornung
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sirisha Achanta
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alison Moss
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - James S. Schwaber
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rajanikanth Vadigepalli
- Department of Pathology and Genomic Medicine, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
12
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
13
|
See EJ, Chaba A, Spano S, Maeda A, Clapham C, Burrell LM, Liu J, Khasin M, Liskaser G, Eastwood G, Bellomo R. Renin Levels and Angiotensin II Responsiveness in Vasopressor-Dependent Hypotension. Crit Care Med 2024; 52:1218-1227. [PMID: 38511994 DOI: 10.1097/ccm.0000000000006273] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
OBJECTIVES The relationship between renin levels, exposure to renin-angiotensin system (RAS) inhibitors, angiotensin II (ANGII) responsiveness, and outcome in patients with vasopressor-dependent vasodilatory hypotension is unknown. DESIGN We conducted a single-center prospective observational study to explore whether recent RAS inhibitor exposure affected baseline renin levels, whether baseline renin levels predicted ANGII responsiveness, and whether renin levels at 24 hours were associated with clinical outcomes. SETTING An academic ICU in Melbourne, VIC, Australia. PATIENTS Forty critically ill adults who received ANGII as the primary agent for vasopressor-dependent vasodilatory hypotension who were included in the Acute Renal effects of Angiotensin II Management in Shock study. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS After multivariable adjustment, recent exposure to a RAS inhibitor was independently associated with a relative increase in baseline renin levels by 198% (95% CI, 36-552%). The peak amount of ANGII required to achieve target mean arterial pressure was independently associated with baseline renin level (increase by 46% per ten-fold increase; 95% CI, 8-98%). Higher renin levels at 24 hours after ANGII initiation were independently associated with fewer days alive and free of continuous renal replacement therapy (CRRT) (-7 d per ten-fold increase; 95% CI, -12 to -1). CONCLUSIONS In patients with vasopressor-dependent vasodilatory hypotension, recent RAS inhibitor exposure was associated with higher baseline renin levels. Such higher renin levels were then associated with decreased ANGII responsiveness. Higher renin levels at 24 hours despite ANGII infusion were associated with fewer days alive and CRRT-free. These preliminary findings emphasize the importance of the RAS and the role of renin as a biomarker in patients with vasopressor-dependent vasodilatory hypotension.
Collapse
Affiliation(s)
- Emily J See
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
- Department of Critical Care, University of Melbourne, Parkville, VIC, Australia
- Australian and New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Intensive Care, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Nephrology, Royal Melbourne Hospital, Parkville, VIC, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
- Institute of Breathing and Sleep, Austin Health, Melbourne, VIC, Australia
- Data Analytics Research and Evaluation Centre, The University of Melbourne and Austin Hospital, Melbourne, VIC, Australia
| | - Anis Chaba
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Sofia Spano
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Akinori Maeda
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Caroline Clapham
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Louise M Burrell
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
- Institute of Breathing and Sleep, Austin Health, Melbourne, VIC, Australia
| | - Jasmine Liu
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Monique Khasin
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Grace Liskaser
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Glenn Eastwood
- Department of Intensive Care, Austin Hospital, Heidelberg, VIC, Australia
| | - Rinaldo Bellomo
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
- Data Analytics Research and Evaluation Centre, The University of Melbourne and Austin Hospital, Melbourne, VIC, Australia
| |
Collapse
|
14
|
McLaughlin L, Zhang B, Sharma S, Knoten AL, Kaushal M, Purkerson JM, Huyck H, Pryhuber GS, Gaut JP, Jain S. Three Dimensional Multiscalar Neurovascular Nephron Connectivity Map of the Human Kidney Across the Lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.29.605633. [PMID: 39211059 PMCID: PMC11361085 DOI: 10.1101/2024.07.29.605633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The human kidney is a vital organ with a remarkable ability to coordinate the activity of up to a million nephrons, its main functional tissue unit (FTU), and maintain homeostasis. We developed tissue processing and analytical methods to construct a 3D map of neurovascular nephron connectivity of the human kidney and glean insights into how this structural organization enables coordination of various functions of the nephron, such as glomerular filtration, solute and water absorption, secretion by the tubules, and regulation of blood flow and pressure by the juxtaglomerular apparatus, in addition to how these functions change across disease and lifespans. Using light sheet fluorescence microscopy (LSFM) and morphometric analysis we discovered changes in anatomical orientation of the vascular pole, glomerular density, volume, and innervation through postnatal development and ageing. The extensive nerve network exists from cortex FTUs to medullary loop of Henle, providing connectivity within segments of the same nephron, and between separate nephrons. The nerves organize glomeruli into discreet communities (in the same network of nerves). Adjacent glomerular communities are connected to intercommunal "mother glomeruli" by nerves, a pattern repeating throughout the cortex. These neuro-nephron networks are not developed in postnatal kidneys and are disrupted in diseased kidneys (diabetic or hydronephrosis). This structural organization likely poises the entire glomerular and juxtaglomerular FTUs to synchronize responses to perturbations in fluid homeostasis, utilizing mother glomeruli as network control centers.
Collapse
|
15
|
Cheval L, Poindessous V, Sampaio JL, Crambert G, Pallet N. Lipidomic Profiling of Kidney Cortical Tubule Segments Identifies Lipotypes with Physiological Implications. FUNCTION 2024; 5:zqae016. [PMID: 38985001 PMCID: PMC11237892 DOI: 10.1093/function/zqae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 07/11/2024] Open
Abstract
A detailed knowledge of the lipid composition of components of nephrons is crucial for understanding physiological processes and the development of kidney diseases. However, the lipidomic composition of kidney tubular segments is unknown. We manually isolated the proximal convoluted tubule (PCT), the cortical thick ascending limb of Henle's loop, and the cortical collecting duct from 5 lean and obese mice and subjected the samples to shotgun lipidomics analysis by high-resolution mass spectrometry acquisition. Across all samples, more than 500 lipid species were identified, quantified, and compared. We observed significant compositional differences among the 3 tubular segments, which serve as true signatures. These intrinsic lipidomic features are associated with a distinct proteomic program that regulates highly specific physiological functions. The distinctive lipidomic features of each of the 3 segments are mostly based on the relative composition of neutral lipids, long-chain polyunsaturated fatty acids, sphingolipids, and ether phospholipids. These features support the hypothesis of a lipotype assigned to specific tubular segments. Obesity profoundly impacts the lipotype of PCT. In conclusion, we present a comprehensive lipidomic analysis of 3 cortical segments of mouse kidney tubules. This valuable resource provides unparalleled detail that enhances our understanding of tubular physiology and the potential impact of pathological conditions.
Collapse
Affiliation(s)
- Lydie Cheval
- Laboratoire de Physiologie Rénale et Tubulopathies, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, 75006 Paris, France
| | - Virginie Poindessous
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, 75015, Paris, France
| | - Julio L Sampaio
- CurieCoreTech Metabolomics and Lipidomics Technology Platform, Institut Curie, 75005, Paris, France
| | - Gilles Crambert
- Laboratoire de Physiologie Rénale et Tubulopathies, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, 75006 Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, 75006 Paris, France
| | - Nicolas Pallet
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, 75015, Paris, France
- Department of Clinical Chemistry, Assistance Publique Hôpitaux de Paris, Georges Pompidou European Hospital, 75015, Paris, France
| |
Collapse
|
16
|
Musah S, Bhattacharya R, Himmelfarb J. Kidney Disease Modeling with Organoids and Organs-on-Chips. Annu Rev Biomed Eng 2024; 26:383-414. [PMID: 38424088 PMCID: PMC11479997 DOI: 10.1146/annurev-bioeng-072623-044010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Kidney disease is a global health crisis affecting more than 850 million people worldwide. In the United States, annual Medicare expenditures for kidney disease and organ failure exceed $81 billion. Efforts to develop targeted therapeutics are limited by a poor understanding of the molecular mechanisms underlying human kidney disease onset and progression. Additionally, 90% of drug candidates fail in human clinical trials, often due to toxicity and efficacy not accurately predicted in animal models. The advent of ex vivo kidney models, such as those engineered from induced pluripotent stem (iPS) cells and organ-on-a-chip (organ-chip) systems, has garnered considerable interest owing to their ability to more accurately model tissue development and patient-specific responses and drug toxicity. This review describes recent advances in developing kidney organoids and organ-chips by harnessing iPS cell biology to model human-specific kidney functions and disease states. We also discuss challenges that must be overcome to realize the potential of organoids and organ-chips as dynamic and functional conduits of the human kidney. Achieving these technological advances could revolutionize personalized medicine applications and therapeutic discovery for kidney disease.
Collapse
Affiliation(s)
- Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA;
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina, USA
- Developmental and Stem Cell Biology Program and Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Rohan Bhattacharya
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, North Carolina, USA;
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, North Carolina, USA
| | - Jonathan Himmelfarb
- Department of Medicine, Kidney Research Institute, and Division of Nephrology, University of Washington School of Medicine, Seattle, Washington, USA;
| |
Collapse
|
17
|
Garcia B, Ter Schiphorst B, Santos K, Su F, Dewachter L, Vasques-Nóvoa F, Rocha-Oliveira E, Roncon-Albuquerque R, Uba T, Hartmann O, Picod A, Azibani F, Callebert J, Goldman S, Annoni F, Favory R, Vincent JL, Creteur J, Taccone FS, Mebazaa A, Herpain A. Inhibition of circulating dipeptidyl-peptidase 3 by procizumab in experimental septic shock reduces catecholamine exposure and myocardial injury. Intensive Care Med Exp 2024; 12:53. [PMID: 38849640 PMCID: PMC11161450 DOI: 10.1186/s40635-024-00638-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Dipeptidyl peptidase 3 (DPP3) is a ubiquitous cytosolic enzyme released into the bloodstream after tissue injury, that can degrade angiotensin II. High concentrations of circulating DPP3 (cDPP3) have been associated with worse outcomes during sepsis. The aim of this study was to assess the effect of Procizumab (PCZ), a monoclonal antibody that neutralizes cDPP3, in an experimental model of septic shock. METHODS In this randomized, open-label, controlled study, 16 anesthetized and mechanically ventilated pigs with peritonitis were randomized to receive PCZ or standard treatment when the mean arterial pressure (MAP) dropped below 50 mmHg. Resuscitation with fluids, antimicrobial therapy, peritoneal lavage, and norepinephrine was initiated one hour later to maintain MAP between 65-75 mmHg for 12 h. Hemodynamic variables, tissue oxygenation indices, and measures of organ failure and myocardial injury were collected. Organ blood flow was assessed using isotopic assessment (99mtechnetium albumin). cDPP3 activity, equilibrium analysis of the renin-angiotensin system and circulating catecholamines were measured. Tissue mRNA expression of interleukin-6 and downregulation of adrenergic and angiotensin receptors were assessed on vascular and myocardial samples. RESULTS PCZ-treated animals had reduced cDPP3 levels and required less norepinephrine and fluid than septic control animals for similar organ perfusion and regional blood flow. PCZ-treated animals had less myocardial injury, and higher PaO2/FiO2 ratios. PCZ was associated with lower circulating catecholamine levels; higher circulating angiotensin II and higher angiotensin II receptor type 1 myocardial protein expression, and with lower myocardial and radial artery mRNA interleukin-6 expression. CONCLUSIONS In an experimental model of septic shock, PCZ administration was associated with reduced fluid and catecholamine requirements, less myocardial injury and cardiovascular inflammation, along with preserved angiotensin II signaling.
Collapse
Affiliation(s)
- Bruno Garcia
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France.
| | - Benoit Ter Schiphorst
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
| | | | - Fuhong Su
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Estela Rocha-Oliveira
- Cardiovascular R&D Center, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Theo Uba
- 4TEEN4 Pharmaceuticals GmbH, Hennigsdorf, Germany
| | | | - Adrien Picod
- Université Paris Cité, UMR-S 942, INSERM, MASCOT, Paris, France
| | - Feriel Azibani
- Université Paris Cité, UMR-S 942, INSERM, MASCOT, Paris, France
| | - Jacques Callebert
- Université Paris Cité, UMR-S 942, INSERM, MASCOT, Paris, France
- Department of Biochemistry, Assistance Publique Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Serge Goldman
- Department of Nuclear Medicine, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Filippo Annoni
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Raphaël Favory
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Jean-Louis Vincent
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jacques Creteur
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fabio Silvio Taccone
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alexandre Mebazaa
- Université Paris Cité, UMR-S 942, INSERM, MASCOT, Paris, France
- Department of Anesthesia, Burn and Critical Care, University Hospitals Saint-Louis-Lariboisière, AP-HP, Paris, France
| | - Antoine Herpain
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Intensive Care, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
18
|
Arfsten H, Heitzinger G, Prausmüller S, Weidenhammer A, Goliasch G, Bartko PE, Spinka G, Hülsmann M, Pavo N. Excess renin is attributed to the combination of forward and backward failure in HFrEF. ESC Heart Fail 2024; 11:1748-1757. [PMID: 38459668 PMCID: PMC11098628 DOI: 10.1002/ehf2.14731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/05/2024] [Accepted: 02/06/2024] [Indexed: 03/10/2024] Open
Abstract
AIMS Regulation of the renin-angiotensin system (RAS) in heart failure (HF) with reduced ejection fraction (HFrEF) still raises questions, as a large proportion of patients show normal renin levels despite manifest disease. Experimental venous congestion results in reduced renal perfusion pressure and stimulates renin secretion. We hypothesized that excess renin levels are mainly a result of right ventricular failure as a sequalae of left ventricular dysfunction. The study aimed to link right ventricular function (RVF) with renin levels and to investigate further contributors to excess RAS activation. METHODS AND RESULTS Three hundred thirty-two chronic HFrEF patients undergoing routine ambulatory care were consecutively enrolled in a prospective, registry-based, observational study. Laboratory parameters, including cardiac-specific markers renin, aldosterone, and N-terminal pro-brain natriuretic peptide (NT-proBNP), echocardiographic examination (n = 247), and right heart catheterization (n = 85), were documented. The relationship between renin and its respective parameters was analysed. Renin concentration was not associated with the New York Heart Association class or NT-proBNP. Systolic blood pressure, systemic vascular resistance, serum sodium, aldosterone, and lactate dehydrogenase were associated with increased renin levels (P < 0.035 for all). Renin levels similarly increased with worsening of RVF parameters such as fractional area change, tricuspid annular plane systolic excursion, tissue Doppler imaging, and inferior vena cava diameter (P < 0.011 for all), but not with pulmonary pressure. Excess renin levels were observed when worsening RVF was combined with reduced renal perfusion {625 μIU/mL [interquartile range (IQR): 182-1761] vs. 67 μIU/mL [IQR: 16-231], P < 0.001}, which was associated with worse survival. CONCLUSIONS While unrelated to classical indices of HF severity, circulating renin levels increase with the worsening of RVF, especially in the combined presence of forward and backward failure. This might explain normal renin levels in HFrEF patients but also excess renin levels in poor haemodynamic conditions.
Collapse
Affiliation(s)
- Henrike Arfsten
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Gregor Heitzinger
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Suriya Prausmüller
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Annika Weidenhammer
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Philipp E. Bartko
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Georg Spinka
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Martin Hülsmann
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of CardiologyMedical University of ViennaWaehringer Guertel 18‐201090ViennaAustria
| |
Collapse
|
19
|
Lobo J, Canete-Portillo S, Pena MDCR, McKenney JK, Aron M, Massicano F, Wilk BM, Gajapathy M, Brown DM, Baydar DE, Matoso A, Rioux-Leclerq N, Pan CC, Tretiakova MS, Trpkov K, Williamson SR, Rais-Bahrami S, Mackinnon AC, Harada S, Worthey EA, Magi-Galluzzi C. Molecular Characterization of Juxtaglomerular Cell Tumors: Evidence of Alterations in MAPK-RAS Pathway. Mod Pathol 2024; 37:100492. [PMID: 38614322 DOI: 10.1016/j.modpat.2024.100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/18/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2024]
Abstract
Juxtaglomerular cell tumor (JGCT) is a rare neoplasm, part of the family of mesenchymal tumors of the kidney. Although the pathophysiological and clinical correlates of JGCT are well known, as these tumors are an important cause of early-onset arterial hypertension refractory to medical treatment, their molecular background is unknown, with only few small studies investigating their karyotype. Herein we describe a multi-institutional cohort of JGCTs diagnosed by experienced genitourinary pathologists, evaluating clinical presentation and outcome, morphologic diversity, and, importantly, the molecular features. Ten JGCTs were collected from 9 institutions, studied by immunohistochemistry, and submitted to whole exome sequencing. Our findings highlight the morphologic heterogeneity of JGCT, which can mimic several kidney tumor entities. Three cases showed concerning histologic features, but the patient course was unremarkable, which suggests that morphologic evaluation alone cannot reliably predict the clinical behavior. Gain-of-function variants in RAS GTPases were detected in JGCTs, with no evidence of additional recurrent genomic alterations. In conclusion, we present the largest series of JGCT characterized by whole exome sequencing, highlighting the putative role of the MAPK-RAS pathway.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto; Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (P.CCC) and RISE@CI-IPOP (Health Research Network), Porto, Portugal; Department of Pathology and Molecular Immunology, ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Sofia Canete-Portillo
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | | | - Jesse K McKenney
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Manju Aron
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Felipe Massicano
- Department of Genetics, Center for Computational Genomics and Data Science, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Brandon M Wilk
- Department of Genetics, Center for Computational Genomics and Data Science, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Manavalan Gajapathy
- Department of Genetics, Center for Computational Genomics and Data Science, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Donna M Brown
- Department of Genetics, Center for Computational Genomics and Data Science, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Dilek E Baydar
- Department of Pathology, Koc University School of Medicine, Istanbul, Turkey
| | - Andres Matoso
- Departments of Pathology, Urology, Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Chin-Chen Pan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Sean R Williamson
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama; Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Alexander C Mackinnon
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Shuko Harada
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Elizabeth A Worthey
- Department of Genetics, Center for Computational Genomics and Data Science, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama
| | - Cristina Magi-Galluzzi
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Albama.
| |
Collapse
|
20
|
Kotani Y, Chappell M, Landoni G, Zarbock A, Bellomo R, Khanna AK. Renin in critically ill patients. Ann Intensive Care 2024; 14:79. [PMID: 38775999 PMCID: PMC11111649 DOI: 10.1186/s13613-024-01304-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
The renin-angiotensin system (RAS) constitutes one of the principal mechanisms to maintain hemodynamic and fluid homeostasis. However, most research until now on RAS primarily focuses on its relationship with hypertension and its role in critically ill hypotensive populations is not well understood. With the approval of angiotensin II (Ang II) in the United States and Europe, following a phase 3 randomized controlled trial showing efficacy in catecholamine-resistant vasodilatory shock, there is growing interest in RAS in critically ill patients. Among the fundamental components of RAS, renin acts as the initial stimulus for the entire system. In the context of hypotension, its release increases in response to low blood pressure sensed by renal baroreceptors and attenuated negative Ang II feedback loop. Thus, elevated renin could reflect disease severity and predict poor outcomes. Studies investigating this hypothesis have validated the prognostic accuracy of renin in various critically ill populations, with several reports indicating its superiority to lactate for mortality prediction. Accordingly, renin reduction has been used to assess the effectiveness of Ang II administration. Furthermore, renin holds potential to identify patients who might benefit from Ang II treatment, potentially paving the way for personalized vasopressor management. Despite these promising data, most available evidence is derived from retrospective analysis and necessitates prospective confirmation. The absence of a rapid, point-of-care and reliable renin assay presents another hurdle to its integration into routine clinical practice. This narrative review aims to describe the current understanding and future directions of renin as a biomarker during resuscitation of critically ill patients.
Collapse
Affiliation(s)
- Yuki Kotani
- Department of Intensive Care Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Mark Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Giovanni Landoni
- Department of Anesthesia and Intensive Care, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Muenster, Muenster, Germany
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Department of Critical Care, Melbourne Medical School, The University of Melbourne, Melbourne, Australia
| | - Ashish K Khanna
- Section On Critical Care Medicine, Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Perioperative Outcomes and Informatics Collaborative, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Outcomes Research Consortium, Cleveland, OH, 44195, USA.
| |
Collapse
|
21
|
Wagner BR, Rao PS. Sodium-glucose cotransporter 2 inhibitors: are they ready for prime time in the management of lupus nephritis? Curr Opin Rheumatol 2024; 36:163-168. [PMID: 38517337 DOI: 10.1097/bor.0000000000001002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
PURPOSE OF REVIEW Lupus nephritis is a common complication of systemic lupus erythematosus and is associated with significant morbidity and mortality. The utility of sodium-glucose cotransporter 2 (SGLT2) inhibitors in the management of lupus nephritis is currently uncertain. Here, we summarize the rationale for their use among patient with lupus nephritis. RECENT FINDINGS SGLT2 inhibitors were initially developed as antihyperglycemic agents. They have since been shown to have additional, profound effects to slow the progression of chronic kidney disease and lessen the long-term risks of cardiovascular disease in large clinic trials of patients with chronic kidney disease, with and without diabetes, as well as in patients with and without proteinuria. Patients with recent exposure to immunosuppression were excluded from these trials due to concern for risk of infection. In the few, small trials of patients with lupus nephritis, SGLT2 inhibitors were found to be well tolerated. They have been shown to reduce proteinuria and to have modest beneficial effects on blood pressure and BMI among patients with lupus nephritis. They have not been shown to influence disease activity. SUMMARY SGLT2 inhibitors may have a role in mitigating the chronic renal and cardiovascular effects of lupus nephritis. They should be introduced after kidney function has been stabilized with appropriate immunosuppression, in conjunction with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers. They currently have no role in active disease.
Collapse
Affiliation(s)
- Benjamin R Wagner
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
22
|
Gyarmati G, Shroff UN, Riquier-Brison A, Desposito D, Ju W, Stocker SD, Izuhara A, Deepak S, Becerra Calderon A, Burford JL, Kadoya H, Moon JY, Chen Y, Rinschen MM, Ahmadi N, Lau L, Biemesderfer D, James AW, Minichiello L, Zlokovic BV, Gill IS, Kretzler M, Peti-Peterdi J. Neuronally differentiated macula densa cells regulate tissue remodeling and regeneration in the kidney. J Clin Invest 2024; 134:e174558. [PMID: 38598837 PMCID: PMC11142747 DOI: 10.1172/jci174558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 04/09/2024] [Indexed: 04/12/2024] Open
Abstract
Tissue regeneration is limited in several organs, including the kidney, contributing to the high prevalence of kidney disease globally. However, evolutionary and physiological adaptive responses and the presence of renal progenitor cells suggest an existing remodeling capacity. This study uncovered endogenous tissue remodeling mechanisms in the kidney that were activated by the loss of body fluid and salt and regulated by a unique niche of a minority renal cell type called the macula densa (MD). Here, we identified neuronal differentiation features of MD cells that sense the local and systemic environment and secrete angiogenic, growth, and extracellular matrix remodeling factors, cytokines and chemokines, and control resident progenitor cells. Serial intravital imaging, MD nerve growth factor receptor and Wnt mouse models, and transcriptome analysis revealed cellular and molecular mechanisms of these MD functions. Human and therapeutic translation studies illustrated the clinical potential of MD factors, including CCN1, as a urinary biomarker and therapeutic target in chronic kidney disease. The concept that a neuronally differentiated key sensory and regulatory cell type responding to organ-specific physiological inputs controls local progenitors to remodel or repair tissues may be applicable to other organs and diverse tissue-regenerative therapeutic strategies.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Urvi Nikhil Shroff
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Anne Riquier-Brison
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Dorinne Desposito
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Audrey Izuhara
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Sachin Deepak
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Alejandra Becerra Calderon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - James L. Burford
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hiroyuki Kadoya
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Ju-Young Moon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California, USA
| | - Markus M. Rinschen
- Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nariman Ahmadi
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Lester Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel Biemesderfer
- Section of Nephrology and Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Inderbir S. Gill
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - János Peti-Peterdi
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
23
|
El Fathi W, van Ochten M, Rehman M, van Kuijk SMJ, IntHout J, Ghossein-Doha C, de Haas S, Spaanderman MEA, van Drongelen J. Active plasma renin concentration throughout healthy and complicated pregnancy: a systematic review and meta-analysis. Reprod Biol Endocrinol 2024; 22:29. [PMID: 38454417 PMCID: PMC10918957 DOI: 10.1186/s12958-024-01200-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Pregnancy is characterized by profound circulatory changes and compensatory adjustments in the renin-angiotensin-aldosterone system (RAAS). Differences in regulatory response may antedate or accompany vascular complicated pregnancy. We performed a systematic review and meta-analysis to delineate the trajectory of active plasma renin concentration (APRC) in healthy pregnancy and compare this to complicated pregnancy. METHODS We performed a systematic review and meta-analysis on APRC during normotensive and hypertensive pregnancies, using PubMed (NCBI) and Embase (Ovid) databases. We included only studies reporting measurements during pregnancy together with a nonpregnant reference group measurement. Risk of bias was assessed with QUIPS. Ratio of the mean (ROM) and 95% confidence intervals (CI) of APRC values between pregnant and nonpregnant women were estimated for predefined intervals of gestational age using a random-effects model. Meta-regression was used to analyze APRC over time. RESULTS In total, we included 18 studies. As compared to nonpregnant, APRC significantly increased as early as the first weeks of healthy pregnancy and stayed increased throughout the whole pregnancy (ROM 2.77; 95% CI 2.26-3.39). APRC in hypertensive complicated pregnancy was not significantly different from nonpregnancy (ROM 1.32; 95% CI 0.97-1.80). CONCLUSION Healthy pregnancy is accompanied by a profound rise in APRC in the first trimester that is maintained until term. In hypertensive complicated pregnancy, this increase in APRC is not observed.
Collapse
Affiliation(s)
- Wisal El Fathi
- Department of Gynecology and Obstetrics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maaike van Ochten
- Department of Gynecology and Obstetrics, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Munieb Rehman
- Department of Cardiology, Isala Hospital, Zwolle, The Netherlands
| | - Sander M J van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joanna IntHout
- Department for Health Evidence, Section Biostatistics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Chahinda Ghossein-Doha
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sander de Haas
- Department of Gynecology and Obstetrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marc E A Spaanderman
- Department of Gynecology and Obstetrics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Gynecology and Obstetrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joris van Drongelen
- Department of Gynecology and Obstetrics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Stocker SD, Kinsman BJ, Farquhar WB, Gyarmati G, Peti-Peterdi J, Sved AF. Physiological Mechanisms of Dietary Salt Sensing in the Brain, Kidney, and Gastrointestinal Tract. Hypertension 2024; 81:447-455. [PMID: 37671571 PMCID: PMC10915107 DOI: 10.1161/hypertensionaha.123.19488] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Excess dietary salt (NaCl) intake is strongly correlated with cardiovascular disease and is a major contributing factor to the pathogenesis of hypertension. NaCl-sensitive hypertension is a multisystem disorder that involves renal dysfunction, vascular abnormalities, and neurogenically-mediated increases in peripheral resistance. Despite a major research focus on organ systems and these effector mechanisms causing NaCl-induced increases in arterial blood pressure, relatively less research has been directed at elucidating how NaCl is sensed by various tissues to elicit these downstream effects. The purpose of this review is to discuss how the brain, kidney, and gastrointestinal tract sense NaCl including key cell types, the role of NaCl versus osmolality, and the underlying molecular and electrochemical mechanisms.
Collapse
Affiliation(s)
- Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine
| | - Brian J Kinsman
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital
| | | | - Georgina Gyarmati
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Janos Peti-Peterdi
- Department of Physiology and Neuroscience and Medicine, Zilkha Neurogenetic Institute, University of Southern California
| | - Alan F. Sved
- Department of Neuroscience, University of Pittsburgh
| |
Collapse
|
25
|
van der Pluijm LA, Koudijs A, Stam W, Roelofs JJ, Danser AJ, Rotmans JI, Gross KW, Pieper MP, van Zonneveld AJ, Bijkerk R. SGLT2 inhibition promotes glomerular repopulation by cells of renin lineage in experimental kidney disease. Acta Physiol (Oxf) 2024; 240:e14108. [PMID: 38314444 PMCID: PMC10923162 DOI: 10.1111/apha.14108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/06/2024]
Abstract
AIM Sodium glucose co-transporter-2 (SGLT2) inhibitors stimulate renal excretion of sodium and glucose and exert renal protective effects in patients with (non-)diabetic chronic kidney disease (CKD) and may as well protect against acute kidney injury (AKI). The mechanism behind this kidney protective effect remains unclear. Juxtaglomerular cells of renin lineage (CoRL) have been demonstrated to function as progenitors for multiple adult glomerular cell types in kidney disease. This study assesses the impact of SGLT2 inhibition on the repopulation of glomerular cells by CoRL and examines their phenotypic commitment. METHODS Experiments were performed in Ren1cre-tdTomato lineage-trace mice. Either 5/6 nephrectomy (5/6NX) modeling CKD or bilateral ischaemia reperfusion injury (bIRI) mimicking AKI was applied, while the SGLT2 inhibitor empagliflozin (10 mg/kg) was administered daily via oral gavage for 14 days. RESULTS Both 5/6NX and bIRI-induced kidney injury increased the number of glomerular CoRL-derived cells. SGLT2 inhibition improved kidney function after 5/6NX, indicated by decreased blood creatinine and urea levels, but not after bIRI. In line with this, empagliflozin in 5/6NX animals resulted in less glomerulosclerosis, while it did not affect histopathological features in bIRI. Treatment with empagliflozin resulted in an increase in the number of CoRL-derived glomerular cells in both 5/6NX and bIRI conditions. Interestingly, SGLT2 inhibition led to more CoRL-derived podocytes in 5/6NX animals, whereas empagliflozin-treated bIRI mice presented with increased levels of parietal epithelial and mesangial cells derived from CoRL. CONCLUSION We conclude that SGLT2 inhibition by empagliflozin promotes CoRL-mediated glomerular repopulation with selective CoRL-derived cell types depending on the type of experimental kidney injury. These findings suggest a previously unidentified mechanism that could contribute to the renoprotective effect of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Loïs A.K. van der Pluijm
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Angela Koudijs
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Joris J.T.H. Roelofs
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - A.H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joris I. Rotmans
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Kenneth W. Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Michael P. Pieper
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| |
Collapse
|
26
|
Martínez-Rojas MÁ, Balcázar H, González-Soria I, González-Rivera JM, Rodríguez-Vergara ME, Velazquez-Villegas LA, León-Contreras JC, Pérez-Villalva R, Correa F, Rosetti F, Bobadilla NA. Transient inhibition of sodium-glucose cotransporter 2 after ischemia/reperfusion injury ameliorates chronic kidney disease. JCI Insight 2024; 9:e173675. [PMID: 38516890 PMCID: PMC11063941 DOI: 10.1172/jci.insight.173675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitor, dapagliflozin (Dapa), exhibited nephroprotective effects in patients with chronic kidney disease (CKD). We assessed the efficacy of short-term Dapa administration following acute kidney injury (AKI) in preventing CKD. Male Wistar rats were randomly assigned to Sham surgery, bilateral ischemia for 30 minutes (abbreviated as IR), and IR + Dapa groups. Daily treatment with Dapa was initiated just 24 hours after IR and maintained for only 10 days. Initially, rats were euthanized at this point to study early renal repair. After severe AKI, Dapa promptly restored creatinine clearance (CrCl) and significantly reduced renal vascular resistance compared with the IR group. Furthermore, Dapa effectively reversed the mitochondrial abnormalities, including increased fission, altered mitophagy, metabolic dysfunction, and proapoptotic signaling. To study this earlier, another set of rats was studied just 5 days after AKI. Despite persistent renal dysfunction, our data reveal a degree of mitochondrial protection. Remarkably, a 10-day treatment with Dapa demonstrated effectiveness in preventing CKD transition in an independent cohort monitored for 5 months after AKI. This was evidenced by improvements in proteinuria, CrCl, glomerulosclerosis, and fibrosis. Our findings underscore the potential of Dapa in preventing maladaptive repair following AKI, emphasizing the crucial role of early intervention in mitigating AKI long-term consequences.
Collapse
Affiliation(s)
- Miguel Ángel Martínez-Rojas
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | - Hiram Balcázar
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | - Isaac González-Soria
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | - Jesús Manuel González-Rivera
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | - Mauricio E. Rodríguez-Vergara
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | | | - Juan Carlos León-Contreras
- Departmento de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Rosalba Pérez-Villalva
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| | - Francisco Correa
- Departmento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Florencia Rosetti
- Departmento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Norma A. Bobadilla
- Unidad de Fisiología Molecular, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departmento de Nefrología y Metabolismo Mineral
| |
Collapse
|
27
|
Stepanova N. SGLT2 inhibitors in peritoneal dialysis: a promising frontier toward improved patient outcomes. RENAL REPLACEMENT THERAPY 2024; 10:5. [DOI: 10.1186/s41100-024-00523-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/14/2024] [Indexed: 01/12/2025] Open
Abstract
AbstractPeritoneal dialysis (PD) stands as an important modality among kidney replacement therapies for end-stage kidney disease, offering patients remarkable flexibility and autonomy. Despite its widespread use, challenges such as glucose-related complications, peritoneal membrane fibrosis, declining renal function, and cardiovascular risks persist, necessitating innovative therapeutic approaches. Sodium–glucose cotransporter 2 (SGLT2) inhibitors, originally developed for treating type 2 diabetes mellitus, have recently shown promise as add-on therapy for patients with diabetic and non-diabetic chronic kidney disease (CKD), even in advanced stages. This review describes the potential role of SGLT2 inhibitors as a breakthrough therapeutic option in PD, emphasizing their ability to address unmet clinical needs and improve patient outcomes. The multiple effects of SGLT2 inhibitors in CKD, including metabolic modulation, antihypertensive, diuretic, anemia-reducing, antioxidant, and antiinflammatory properties, are reviewed in the context of PD challenges. Additionally, the potentially protective influence of SGLT2 inhibitors on the integrity of the peritoneal membrane and the transport of solutes and water in the peritoneum are emphasized. Despite these encouraging results, the paper highlights the potential risks associated with SGLT2 inhibitors in PD and emphasizes the need for cautious and thorough investigation of dosing, long-term safety considerations, and patient-specific factors through comprehensive clinical trials. Looking forward, the review argues for well-designed studies to evaluate the expanded safety profile of SGLT2 inhibitors in PD, with particular attention paid to peritoneal membrane integrity and overall patient outcomes.
Collapse
|
28
|
Kömhoff M, Gracchi V, Dijkman H, Beck BB, Monnens L. Hyporeninemic hypoaldosteronism in RMND1-related mitochondrial disease. Pediatr Nephrol 2024; 39:125-129. [PMID: 37450011 PMCID: PMC10673983 DOI: 10.1007/s00467-023-06079-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND RMND1 is a nuclear gene needed for proper function of mitochondria. A pathogenic gene will cause multiple oxidative phosphorylation defects. A renal phenotype consisting of hyponatremia, hyperkalemia, and acidosis is frequently reported, previously considered to be due to aldosterone insensitivity. METHODS Clinical features and pathophysiology of three patients will be reported. DNA of these patients was subjected to exome screening. RESULTS In the first family, one pathogenic heterozygous and one highly probable heterozygous mutation were detected. In the second family, a homozygous pathogenic mutation was present. The electrolyte disbalance was not due to aldosterone insensitivity but to low plasma aldosterone concentration, a consequence of low plasma renin activity. This disbalance can be treated. In all three patients, the kidney function declined. In the first family, both children suffered from an unexplained arterial thrombosis with dire consequences. CONCLUSIONS Hyporeninemic hypoaldosteronism is the mechanism causing the electrolyte disbalance reported in patients with RMND1 mutations, and can be treated.
Collapse
Affiliation(s)
- Martin Kömhoff
- University Children's Hospital, Philipps University, Marburg, Germany
| | - Valentina Gracchi
- Department of Pediatrics, UMCG, University Groningen, Groningen, the Netherlands
| | - Henry Dijkman
- Department of Pathology, Radboud University Centre, Nijmegen, the Netherlands
| | - Bodo B Beck
- Department of Human Genetics, Cologne, Germany
| | - Leo Monnens
- Department of Physiology, Radboud University Centre, Nijmegen, the Netherlands.
| |
Collapse
|
29
|
Rai B, Srivastava J, Saxena P. The Functional Role of microRNAs and mRNAs in Diabetic Kidney Disease: A Review. Curr Diabetes Rev 2024; 20:e201023222412. [PMID: 37867275 DOI: 10.2174/0115733998270983231009094216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 10/24/2023]
Abstract
Diabetes is a group of diseases marked by poor control of blood glucose levels. Diabetes mellitus (DM) occurs when pancreatic cells fail to make insulin, which is required to keep blood glucose levels stable, disorders, and so on. High glucose levels in the blood induce diabetic effects, which can cause catastrophic damage to bodily organs such as the eyes and lower extremities. Diabetes is classified into many forms, one of which is controlled by hyperglycemia or Diabetic Kidney Disease (DKD), and another that is not controlled by hyperglycemia (nondiabetic kidney disease or NDKD) and is caused by other factors such as hypertension, hereditary. DKD is associated with diabetic nephropathy (DN), a leading cause of chronic kidney disease (CKD) and end-stage renal failure. The disease is characterized by glomerular basement membrane thickening, glomerular sclerosis, and mesangial expansion, resulting in a progressive decrease in glomerular filtration rate, glomerular hypertension, and renal failure or nephrotic syndrome. It is also represented by some microvascular complications such as nerve ischemia produced by intracellular metabolic changes, microvascular illness, and the direct impact of excessive blood glucose on neuronal activity. Therefore, DKD-induced nephrotic failure is worse than NDKD. MicroRNAs (miRNAs) are important in the development and progression of several diseases, including diabetic kidney disease (DKD). These dysregulated miRNAs can impact various cellular processes, including inflammation, fibrosis, oxidative stress, and apoptosis, all of which are implicated during DKD. MiRNAs can alter the course of DKD by targeting several essential mechanisms. Understanding the miRNAs implicated in DKD and their involvement in disease development might lead to identifying possible therapeutic targets for DKD prevention and therapy. Therefore, this review focuses specifically on DKD-associated DN, as well as how in-silico approaches may aid in improving the management of the disease.
Collapse
Affiliation(s)
- Bhuvnesh Rai
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Jyotika Srivastava
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Pragati Saxena
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
30
|
Garcia B, Zarbock A, Bellomo R, Legrand M. The role of renin-angiotensin system in sepsis-associated acute kidney injury: mechanisms and therapeutic implications. Curr Opin Crit Care 2023; 29:607-613. [PMID: 37861190 DOI: 10.1097/mcc.0000000000001092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
PURPOSE OF REVIEW This review aims to explore the relationship between the renin angiotensin system (RAS) and sepsis-associated acute kidney injury (SA-AKI), a common complication in critically ill patients associated with mortality, morbidity, and long-term cardiovascular complications. Additionally, this review aims to identify potential therapeutic approaches to intervene with the RAS and prevent the development of AKI. RECENT FINDINGS Recent studies have provided increasing evidence of RAS alteration during sepsis, with systemic and local RAS disturbance, which can contribute to SA-AKI. Angiotensin II was recently approved for catecholamine resistant vasodilatory shock and has been associated with improved outcomes in selected patients. SUMMARY SA-AKI is a common condition that can involve disturbances in the RAS, particularly the canonical angiotensin-converting enzyme (ACE) angiotensin-II (Ang II)/angiotensin II receptor 1 (AT-1R) axis. Increased renin levels, a key enzyme in the RAS, have been shown to be associated with AKI and may also guide vasopressor therapy in shock. In patients with high renin levels, angiotensin II administration may reduce renin concentration, improve intra-renal hemodynamics, and enhance signaling through the angiotensin II receptor 1. Further studies are needed to explore the role of the RAS in SA-AKI and the potential for targeted therapies.
Collapse
Affiliation(s)
- Bruno Garcia
- Department of Anesthesia & Peri-operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
- Experimental Laboratory of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Münster, Germany
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital
- Australian and New Zealand Intensive Care Research Centre, Monash University
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
| | - Matthieu Legrand
- Department of Anesthesia & Peri-operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| |
Collapse
|
31
|
Garcia B, Zarbock A, Bellomo R, Legrand M. The alternative renin-angiotensin system in critically ill patients: pathophysiology and therapeutic implications. Crit Care 2023; 27:453. [PMID: 37986086 PMCID: PMC10662652 DOI: 10.1186/s13054-023-04739-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023] Open
Abstract
The renin-angiotensin system (RAS) plays a crucial role in regulating blood pressure and the cardio-renal system. The classical RAS, mainly mediated by angiotensin I, angiotensin-converting enzyme, and angiotensin II, has been reported to be altered in critically ill patients, such as those in vasodilatory shock. However, recent research has highlighted the role of some components of the counterregulatory axis of the classical RAS, termed the alternative RAS, such as angiotensin-converting Enzyme 2 (ACE2) and angiotensin-(1-7), or peptidases which can modulate the RAS like dipeptidyl-peptidase 3, in many critical situations. In cases of shock, dipeptidyl-peptidase 3, an enzyme involved in the degradation of angiotensin and opioid peptides, has been associated with acute kidney injury and mortality and preclinical studies have tested its neutralization. Angiotensin-(1-7) has been shown to prevent septic shock development and improve outcomes in experimental models of sepsis. In the context of experimental acute lung injury, ACE2 activity has demonstrated a protective role, and its inactivation has been associated with worsened lung function, leading to the use of active recombinant human ACE2, in preclinical and human studies. Angiotensin-(1-7) has been tested in experimental models of acute lung injury and in a recent randomized controlled trial for patients with COVID-19 related hypoxemia. Overall, the alternative RAS appears to have a role in the pathogenesis of disease in critically ill patients, and modulation of the alternative RAS may improve outcomes. Here, we review the available evidence regarding the methods of analysis of the RAS, pathophysiological disturbances of this system, and discuss how therapeutic manipulation may improve outcomes in the critically ill.
Collapse
Affiliation(s)
- Bruno Garcia
- Department of Anesthesia and Peri-Operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Department of Intensive Care, Centre Hospitalier Universitaire de Lille, Lille, France
- Experimental Laboratory of the Department of Intensive Care, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Münster, Germany
| | - Rinaldo Bellomo
- Department of Intensive Care, Austin Hospital, Melbourne, VIC, 3084, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University, Melbourne, Australia
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
| | - Matthieu Legrand
- Department of Anesthesia and Peri-Operative Care, Division of Critical Care Medicine, University of California, San Francisco (UCSF), San Francisco, CA, USA.
| |
Collapse
|
32
|
Dugbartey GJ. Physiological role of hydrogen sulfide in the kidney and its therapeutic implications for kidney diseases. Biomed Pharmacother 2023; 166:115396. [PMID: 37647689 DOI: 10.1016/j.biopha.2023.115396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/21/2023] [Accepted: 08/26/2023] [Indexed: 09/01/2023] Open
Abstract
For over three centuries, hydrogen sulfide (H2S) has been known as a toxic and deadly gas at high concentrations, with a distinctive smell of rotten eggs. However, studies over the past two decades have shown that H2S has risen above its historically notorious label and has now received significant scientific attention as an endogenously produced gaseous signaling molecule that participates in cellular homeostasis and influences a myriad of physiological and pathological processes at low concentrations. Its endogenous production is enzymatically regulated, and when dysregulated, contributes to pathogenesis of renal diseases. In addition, exogenous H2S administration has been reported to exhibit important therapeutic characteristics that target multiple molecular pathways in common renal pathologies in which reduced levels of renal and plasma H2S were observed. This review highlights functional anatomy of the kidney and renal production of H2S. The review also discusses current understanding of H2S in renal physiology and seeks to lay the foundation as a new targeted therapeutic agent for renal pathologies such as hypertensive nephropathy, diabetic kidney disease and water balance disorders.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
33
|
Gronda E, Palazzuoli A, Iacoviello M, Benevenuto M, Gabrielli D, Arduini A. Renal Oxygen Demand and Nephron Function: Is Glucose a Friend or Foe? Int J Mol Sci 2023; 24:9957. [PMID: 37373108 PMCID: PMC10298324 DOI: 10.3390/ijms24129957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
The kidneys and heart work together to balance the body's circulation, and although their physiology is based on strict inter dependence, their performance fulfills different aims. While the heart can rapidly increase its own oxygen consumption to comply with the wide changes in metabolic demand linked to body function, the kidneys physiology are primarily designed to maintain a stable metabolic rate and have a limited capacity to cope with any steep increase in renal metabolism. In the kidneys, glomerular population filters a large amount of blood and the tubular system has been programmed to reabsorb 99% of filtrate by reabsorbing sodium together with other filtered substances, including all glucose molecules. Glucose reabsorption involves the sodium-glucose cotransporters SGLT2 and SGLT1 on the apical membrane in the proximal tubular section; it also enhances bicarbonate formation so as to preserve the acid-base balance. The complex work of reabsorption in the kidney is the main factor in renal oxygen consumption; analysis of the renal glucose transport in disease states provides a better understanding of the renal physiology changes that occur when clinical conditions alter the neurohormonal response leading to an increase in glomerular filtration pressure. In this circumstance, glomerular hyperfiltration occurs, imposing a higher metabolic demand on kidney physiology and causing progressive renal impairment. Albumin urination is the warning signal of renal engagement over exertion and most frequently heralds heart failure development, regardless of disease etiology. The review analyzes the mechanisms linked to renal oxygen consumption, focusing on sodium-glucose management.
Collapse
Affiliation(s)
- Edoardo Gronda
- Medicine and Medicine Sub-Specialties Department, Cardio Renal Program, U.O.C. Nephrology, Dialysis and Adult Renal Transplant Program, IRCCS Ca’ Granda Foundation, Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio Thoracic and Vascular Department, S. Maria alle Scotte Hospital University of Siena, 53100 Siena, Italy;
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
| | - Manuela Benevenuto
- Unità Operativa Complessa Cardiologia-UTIC-Emodinamica, PO Giuseppe Mazzini, 64100 Teramo, Italy;
| | - Domenico Gabrielli
- Unità Operativa Complessa Cardiologia-UTIC, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy;
| | | |
Collapse
|
34
|
Tossetta G, Fantone S, Senzacqua M, Galosi AB, Marzioni D, Morroni M. ZO-1 expression in normal human macula densa: Immunohistochemical and immunofluorescence investigations. J Anat 2023; 242:1184-1188. [PMID: 36719664 PMCID: PMC10184539 DOI: 10.1111/joa.13832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 02/01/2023] Open
Abstract
The macula densa (MD) is an anatomical structure having a plaque shape, placed in the distal end of thick ascending limb of each nephron and belonging to juxtaglomerular apparatus (JGA). The aim of the present investigation is to investigate the presence of ZO-1, a specific marker of tight juncions (TJs), in MD cells. Six samples of normal human renal tissue were embedded in paraffin for ZO-1 expression analysis by immunohistochemical and immunofluorescence techniques. We detected ZO-1 expression in the apical part of cell membrane in MD cells by immunohistochemistry. In addition, ZO-1 and nNOS expressions (a specific marker of MD) were colocalized in MD cells providing clear evidence of TJs presence in normal human MD. Since ZO-1 is responsible for diffusion barrier formation, its presence in the MD supports the existence of a tubulomesangial barrier that ensures a regulated exchange between MD and JGA effectors in renal and glomerular haemodynamic homeostasis.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, School of MedicineUniversità Politecnica delle MarcheAnconaItaly
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, School of MedicineUniversità Politecnica delle MarcheAnconaItaly
| | - Martina Senzacqua
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, School of MedicineUniversità Politecnica delle MarcheAnconaItaly
| | - Andrea Benedetto Galosi
- Division of Urology, Department of Clinical and Specialist SciencesUniversità Politecnica delle Marche, Azienda Ospedaliero‐Universitaria Ospedali RiunitiAnconaItaly
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, School of MedicineUniversità Politecnica delle MarcheAnconaItaly
| | - Manrico Morroni
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, School of MedicineUniversità Politecnica delle MarcheAnconaItaly
- Electron Microscopy UnitAzienda Ospedaliero‐Universitaria Ospedali RiunitiAnconaItaly
| |
Collapse
|
35
|
Cai L, Wang D, Gui T, Wang X, Zhao L, Boron WF, Chen LM, Liu Y. Dietary sodium enhances the expression of SLC4 family transporters, IRBIT, L-IRBIT, and PP1 in rat kidney: Insights into the molecular mechanism for renal sodium handling. Front Physiol 2023; 14:1154694. [PMID: 37082243 PMCID: PMC10111226 DOI: 10.3389/fphys.2023.1154694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
The kidney plays a central role in maintaining the fluid and electrolyte homeostasis in the body. Bicarbonate transporters NBCn1, NBCn2, and AE2 are expressed at the basolateral membrane of the medullary thick ascending limb (mTAL). In a previous study, NBCn1, NBCn2, and AE2 are proposed to play as a regulatory pathway to decrease NaCl reabsorption in the mTAL under high salt condition. When heterologously expressed, the activity of these transporters could be stimulated by the InsP3R binding protein released with inositol 1,4,5-trisphosphate (IRBIT), L-IRBIT (collectively the IRBITs), or protein phosphatase PP1. In the present study, we characterized by immunofluorescence the expression and localization of the IRBITs, and PP1 in rat kidney. Our data showed that the IRBITs were predominantly expressed from the mTAL through the distal renal tubules. PP1 was predominantly expressed in the TAL, but is also present in high abundance from the distal convoluted tubule through the medullary collecting duct. Western blotting analyses showed that the abundances of NBCn1, NBCn2, and AE2 as well as the IRBITs and PP1 were greatly upregulated in rat kidney by dietary sodium. Co-immunoprecipitation study provided the evidence for protein interaction between NBCn1 and L-IRBIT in rat kidney. Taken together, our data suggest that the IRBITs and PP1 play an important role in sodium handling in the kidney. We propose that the IRBITs and PP1 stimulates NBCn1, NBCn2, and AE2 in the basolateral mTAL to inhibit sodium reabsorption under high sodium condition. Our study provides important insights into understanding the molecular mechanism for the regulation of sodium homeostasis in the body.
Collapse
Affiliation(s)
- Lu Cai
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dengke Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Tianxiang Gui
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoyu Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingyu Zhao
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Walter F. Boron
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Li-Ming Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Li-Ming Chen, ; Ying Liu,
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, School of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Li-Ming Chen, ; Ying Liu,
| |
Collapse
|
36
|
Koirala A, Pourafshar N, Daneshmand A, Wilcox CS, Mannemuddhu SS, Arora N. Etiology and Management of Edema: A Review. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:110-123. [PMID: 36868727 DOI: 10.1053/j.akdh.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 04/18/2023]
Abstract
The development of peripheral edema can often pose a significant diagnostic and therapeutic challenge for practitioners due to its association with a wide variety of underlying disorders ranging in severity. Updates to the original Starling's principle have provided new mechanistic insights into edema formation. Additionally, contemporary data highlighting the role of hypochloremia in the development of diuretic resistance provide a possible new therapeutic target. This article reviews the pathophysiology of edema formation and discusses implications for treatment.
Collapse
Affiliation(s)
- Abbal Koirala
- Division of Nephrology, University of Washington, Seattle, WA
| | - Negiin Pourafshar
- Division of Nephrology, MedStar Georgetown University Hospital, Washington DC
| | - Arvin Daneshmand
- Division of Nephrology, MedStar Georgetown University Hospital, Washington DC
| | | | | | - Nayan Arora
- Division of Nephrology, University of Washington, Seattle, WA.
| |
Collapse
|
37
|
Van Beusecum JP, Rianto F, Teakell J, Kon V, Sparks MA, Hoorn EJ, Kirabo A, Ramkumar N. Novel Concepts in Nephron Sodium Transport: A Physiological and Clinical Perspective. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:124-136. [PMID: 36868728 DOI: 10.1053/j.akdh.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 04/13/2023]
Abstract
The kidneys play a critical role in maintaining total body sodium (Na+) balance across a wide range of dietary intake, accomplished by a concerted effort involving multiple Na+ transporters along the nephron. Furthermore, nephron Na+ reabsorption and urinary Na+ excretion are closely linked to renal blood flow and glomerular filtration such that perturbations in either of them can modify Na+ transport along the nephron, ultimately resulting in hypertension and other Na+-retentive states. In this article, we provide a brief physiological overview of nephron Na+ transport and illustrate clinical syndromes and therapeutic agents that affect Na+ transporter function. We highlight recent advances in kidney Na+ transport, particularly the role of immune cells, lymphatics, and interstitial Na+ in regulating Na+ reabsorption, the emergence of potassium (K+) as a regulator of Na+ transport, and the evolution of the nephron to modulate Na+ transport.
Collapse
Affiliation(s)
- Justin P Van Beusecum
- Ralph H. Johnson VA Medical Center, Charleston, SC; Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC
| | - Fitra Rianto
- Division of Nephrology, Department of Medicine, Duke University School of Medicine and Renal Section, Durham VA Health Care System Durham, Durham, NC
| | - Jade Teakell
- Division of Renal Diseases and Hypertension, Department of Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, TX
| | - Valentina Kon
- Division of Nephrology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine and Renal Section, Durham VA Health Care System Durham, Durham, NC
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Nirupama Ramkumar
- Division of Nephrology and Hypertension, Department of Medicine, University of Utah Health, Salt Lake City, UT.
| |
Collapse
|
38
|
Wang Y, Mao X, Shi S, Xu X, Lv J, Zhang B, Wu H, Song Q. SGLT2 inhibitors in the treatment of type 2 cardiorenal syndrome: Focus on renal tubules. FRONTIERS IN NEPHROLOGY 2023; 2:1109321. [PMID: 37674989 PMCID: PMC10479647 DOI: 10.3389/fneph.2022.1109321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/22/2022] [Indexed: 09/08/2023]
Abstract
The pathogenesis of type 2 cardiorenal syndrome (CRS) is mostly associated with reduced cardiac output, increased central venous pressure (CVP), activation of the renin-angiotensin-aldosterone system (RAAS), inflammation, and oxidative stress. As a drug to treat diabetes, sodium-glucose transporter 2 inhibitor (SGLT2i) has been gradually found to have a protective effect on the heart and kidney and has a certain therapeutic effect on CRS. In the process of chronic heart failure (CHF) leading to chronic renal insufficiency, the renal tubular system, as the main functional part of the kidney, is the first to be damaged, but this damage can be reversed. In this review, we focus on the protective mechanisms of SGLT2i targeting renal tubular in the treatment of CRS, including natriuresis and diuresis to relieve renal congestion, attenuate renal tubular fibrosis, improve energy metabolism of renal tubular, and slow tubular inflammation and oxidative stress. This may have beneficial effects on the treatment of CRS and is a direction for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingqiao Song
- Guang ‘anmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
39
|
Kazory A. Chloride and Cardiorenal Interactions in Heart Failure. Nephron Clin Pract 2023; 147:6-8. [PMID: 35640556 DOI: 10.1159/000524987] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022] Open
Abstract
The cardiorenal literature has long been dominated by a sodium-centric view. However, mechanisms affecting sodium homeostasis in patients with heart failure (HF) commonly lead to concurrent changes in the serum levels of chloride as well. There is a growing body of evidence on a strong link between low serum chloride levels and adverse outcomes in HF, which might be even more potent than that of sodium. Maladaptive neurohormonal activation and unresponsiveness to diuretics have been proposed as potential mechanisms to explain this phenomenon. In parallel with accumulating evidence on the predictive value of chloride in various HF populations, the limited available interventional studies that were aimed at increasing serum chloride levels have also shown promising results. Ongoing studies are designed to elucidate the role of chloride as a key cardiorenal connector and whether hypochloremia represents a modifiable risk factor (i.e., target of therapy) or a mere marker of disease severity and poor prognosis.
Collapse
Affiliation(s)
- Amir Kazory
- Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
40
|
Emerging mechanisms involving brain Kv7 channel in the pathogenesis of hypertension. Biochem Pharmacol 2022; 206:115318. [PMID: 36283445 DOI: 10.1016/j.bcp.2022.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Hypertension is a prevalent health problem inducing many organ damages. The pathogenesis of hypertension involves a complex integration of different organ systems including the brain. The elevated sympathetic nerve activity is closely related to the etiology of hypertension. Ion channels are critical regulators of neuronal excitability. Several mechanisms have been proposed to contribute to hypothalamic-driven elevated sympathetic activity, including altered ion channel function. Recent findings indicate one of the voltage-gated potassium channels, Kv7 channels (M channels), plays a vital role in regulating cardiovascular-related neurons activity, and the expression of Kv7 channels is downregulated in hypertension. This review highlights recent findings that the Kv7 channels in the brain, blood vessels, and kidneys are emerging targets involved in the pathogenesis of hypertension, suggesting new therapeutic targets for treating drug-resistant, neurogenic hypertension.
Collapse
|
41
|
Schnell J, Achieng M, Lindström NO. Principles of human and mouse nephron development. Nat Rev Nephrol 2022; 18:628-642. [PMID: 35869368 DOI: 10.1038/s41581-022-00598-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 12/17/2022]
Abstract
The mechanisms underlying kidney development in mice and humans is an area of intense study. Insights into kidney organogenesis have the potential to guide our understanding of the origin of congenital anomalies and enable the assembly of genetic diagnostic tools. A number of studies have delineated signalling nodes that regulate positional identities and cell fates of nephron progenitor and precursor cells, whereas cross-species comparisons have markedly enhanced our understanding of conserved and divergent features of mammalian kidney organogenesis. Greater insights into the complex cellular movements that occur as the proximal-distal axis is established have challenged our understanding of nephron patterning and provided important clues to the elaborate developmental context in which human kidney diseases can arise. Studies of kidney development in vivo have also facilitated efforts to recapitulate nephrogenesis in kidney organoids in vitro, by providing a detailed blueprint of signalling events, cell movements and patterning mechanisms that are required for the formation of correctly patterned nephrons and maturation of physiologically functional apparatus that are responsible for maintaining human health.
Collapse
Affiliation(s)
- Jack Schnell
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA
| | - MaryAnne Achieng
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA
| | - Nils Olof Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
42
|
Scattered Tubular Cells Markers in Macula Densa of Normal Human Adult Kidney. Int J Mol Sci 2022; 23:ijms231810504. [PMID: 36142420 PMCID: PMC9500602 DOI: 10.3390/ijms231810504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/28/2022] Open
Abstract
Background: The scattered tubular cells (STCs) are a population of resident progenitor tubular cells with expansion, self-renewal and epithelial differentiation abilities. Although these cells are localized within the proximal (PTs) and distal (DTs) tubules in a normal adult kidney, their presence has never been demonstrated in human macula densa (MD). The purpose of the present study is to describe the presence of STCs in MD using specific markers such as prominin-1 (CD133), cytokeratin 7 (KRT7) and vimentin (VIM). Methods: We analyzed two sets of three consecutive serial sections for each sample. The first sections of each set were immunostained for nNOS to identify MD, the second sections were immune-stained for CD133 (specific STCs marker) while the third sections were analyzed for KRT7 (another STCs specific marker) and VIM (that stains the basal pole of the STCs) in the first and second sets, respectively, in order to study the co-expression of KRT7 and VIM with the CD133 marker. Results: CD133 was localized in some MD cells and in the adjacent DT cells. Moreover, CD133 was detected in the parietal epithelial cells of Bowman’s capsule and in some proximal tubules (PT). KRT7-positive cells were identified in MD and adjacent DT cells, while KRT7 positivity was mostly confined in both DT and collecting ducts (CD) in the other areas of the renal parenchyma. CD133 and KRT7 were co-expressed in some MD and adjacent DT cells. Some of the latter cells were positive both for CD133 and VIM. CD133 was always localized in the apical part of the cells, whereas the VIM expression was evident only in the cellular basal pole. Although some cells of MD expressed VIM or CD133, none of them co-expressed VIM and CD133. Conclusions: The presence of STCs was demonstrated in human adult MD, suggesting that this structure has expansion, self-renewal and epithelial differentiation abilities, similar to all other parts of renal tubules.
Collapse
|
43
|
Ho KM, Morgan DJR. The Proximal Tubule as the Pathogenic and Therapeutic Target in Acute Kidney Injury. Nephron Clin Pract 2022; 146:494-502. [PMID: 35272287 DOI: 10.1159/000522341] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/27/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In 2004, the term acute kidney injury (AKI) was introduced with the intention of broadening our understanding of rapid declines in renal function and to replace the historical terms of acute renal failure and acute tubular necrosis (ATN). Despite this evolution in terminology, the mechanisms of AKI have stayed largely elusive with the pathophysiological concepts of ATN remaining the mainstay in our understanding of AKI. SUMMARY The proximal tubule (PT), having the highest mitochondrial content in the kidney and relying heavily on oxidative phosphorylation to generate ATP, is vulnerable to ischaemic insults and mitochondrial dysfunction. Histologically, pathological changes in the PT are more consistent than changes to the glomeruli or the loop of Henle in AKI. Physiologically, activation of tubuloglomerular feedback due to PT dysfunction leads to an increase in preglomerular afferent arteriole resistance and a reduction in glomerular filtration. Pharmacologically, frusemide - a drug commonly used in the setting of oliguric AKI - is actively secreted by the PT and its diuretic effect is compromised by its failure to be secreted into the urine and thus be delivered to its site of action at the loop of Henle in AKI. Increases in the urinary, but not plasma biomarkers, of PT injury within 1 h of shock suggest that the PT as the initiation pathogenic target of AKI. KEY MESSAGE Therapeutic agents targeting specifically the PT epithelial cells, in particular its mitochondria - including amino acid ergothioneine and superoxide scavenger MitoTEMPO - show great promises in ameliorating AKI.
Collapse
Affiliation(s)
- Kwok M Ho
- Department of Intensive Care Medicine, Royal Perth Hospital, Perth, Washington, Australia.,Medical School, University of Western Australia, Perth, Washington, Australia.,School of Veterinary & Life Sciences, Murdoch University, Perth, Washington, Australia
| | - David J R Morgan
- Department of Intensive Care Medicine, Fiona Stanley Hospital, Perth, Washington, Australia
| |
Collapse
|
44
|
Onoda N, Kawabata A, Hasegawa K, Sakakura M, Urakawa I, Seki M, Zenkoh J, Suzuki A, Suzuki Y. Spatial and single-cell transcriptome analysis reveals changes in gene expression in response to drug perturbation in rat kidney. DNA Res 2022; 29:dsac007. [PMID: 35325072 PMCID: PMC9014450 DOI: 10.1093/dnares/dsac007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/22/2022] [Indexed: 12/25/2022] Open
Abstract
The kidney is a complex organ that consists of various types of cells. It is occasionally difficult to resolve molecular alterations and possible perturbations that the kidney experiences due to drug-induced damage. In this study, we performed spatial and single-cell transcriptome analysis of rat kidneys and constructed a precise rat renal cell atlas with spatial information. Using the constructed catalogue, we were able to characterize cells of several minor populations, such as macula densa or juxtaglomerular cells. Further inspection of the spatial gene expression data allowed us to identify the upregulation of genes involved in the renin regulating pathway in losartan-treated populations. Losartan is an angiotensin II receptor antagonist drug, and the observed upregulation of the renin pathway-related genes could be due to feedback from the hypotensive action of the drug. Furthermore, we found spatial heterogeneity in the response to losartan among the glomeruli. These results collectively indicate that integrated single-cell and spatial gene expression analysis is a powerful approach to reveal the detailed associations between the different cell types spanning the complicated renal compartments.
Collapse
Affiliation(s)
- Naoki Onoda
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-0882, Japan
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida-shi, Tokyo 194-8533, Japan
| | - Ayako Kawabata
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida-shi, Tokyo 194-8533, Japan
| | - Kumi Hasegawa
- Biomedical Science Research Laboratories 1, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida-shi, Tokyo 194-8533, Japan
| | - Megumi Sakakura
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida-shi, Tokyo 194-8533, Japan
| | - Itaru Urakawa
- Research Core Function Laboratories, Research Unit, R&D Division, Kyowa Kirin Co., Ltd., Machida-shi, Tokyo 194-8533, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-0882, Japan
| | - Junko Zenkoh
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-0882, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-0882, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa-shi, Chiba 277-0882, Japan
| |
Collapse
|
45
|
Mukherjee T, Behl T, Sharma S, Sehgal A, Singh S, Sharma N, Mathew B, Kaur J, Kaur R, Das M, Aleya L, Bungau S. Anticipated pharmacological role of Aviptadil on COVID-19. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:8109-8125. [PMID: 34846667 PMCID: PMC8630992 DOI: 10.1007/s11356-021-17824-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/24/2021] [Indexed: 04/16/2023]
Abstract
Vasoactive intestinal peptide (VIP) is a neuropeptide that is produced by the lymphoid cells and plays a major role in immunological functions for controlling the homeostasis of the immune system. VIP has been identified as a potent anti-inflammatory factor, in boosting both innate and adaptive immunity. Since December 2019, SARS-Cov-2 was found responsible for the disease COVID-19 which has spread worldwide. No specific therapies or 100% effective vaccines are yet available for the treatment of COVID-19. Drug repositioning may offer a strategy and several drugs have been repurposed, including lopinavir/ritonavir, remdesivir, favipiravir, and tocilizumab. This paper describes the main pharmacological properties of synthetic VIP drug (Aviptadil) which is now under clinical trials. A patented formulation of vasoactive intestinal polypeptide (VIP), named RLF-100 (Aviptadil), was developed and finally got approved for human trials by FDA in 2001 and in European medicines agency in 2005. It was awarded Orphan Drug Designation in 2001 by the US FDA for the treatment of acute respiratory distress syndrome and for the treatment of pulmonary arterial hypertension in 2005. Investigational new drug (IND) licenses for human trials of Aviptadil was guaranteed by both the US FDA and EMEA. Preliminary clinical trials seem to support Aviptadil's benefit. However, such drugs like Aviptadil in COVID-19 patients have peculiar safety profiles. Thus, adequate clinical trials are necessary for these compounds.
Collapse
Affiliation(s)
- Tuhin Mukherjee
- Guru Nanak Institute of Pharmaceutical Science and Technology, Kolkata, West Bengal, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Sanchay Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Kochi, India
| | - Jasleen Kaur
- Guru Nanak Institute of Pharmaceutical Science and Technology, Kolkata, West Bengal, India
| | - Ratandeep Kaur
- Guru Nanak Institute of Pharmaceutical Science and Technology, Kolkata, West Bengal, India
| | - Mayukh Das
- Guru Nanak Institute of Pharmaceutical Science and Technology, Kolkata, West Bengal, India
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
46
|
Tsilosani A, Gao C, Zhang W. Aldosterone-Regulated Sodium Transport and Blood Pressure. Front Physiol 2022; 13:770375. [PMID: 35197862 PMCID: PMC8859437 DOI: 10.3389/fphys.2022.770375] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aldosterone is a major mineralocorticoid steroid hormone secreted by glomerulosa cells in the adrenal cortex. It regulates a variety of physiological responses including those to oxidative stress, inflammation, fluid disruption, and abnormal blood pressure through its actions on various tissues including the kidney, heart, and the central nervous system. Aldosterone synthesis is primarily regulated by angiotensin II, K+ concentration, and adrenocorticotrophic hormone. Elevated serum aldosterone levels increase blood pressure largely by increasing Na+ re-absorption in the kidney through regulating transcription and activity of the epithelial sodium channel (ENaC). This review focuses on the signaling pathways involved in aldosterone synthesis and its effects on Na+ reabsorption through ENaC.
Collapse
Affiliation(s)
- Akaki Tsilosani
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Chao Gao
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Wenzheng Zhang
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
47
|
Shroff UN, Gyarmati G, Izuhara A, Deepak S, Peti-Peterdi J. A new view of macula densa cell protein synthesis. Am J Physiol Renal Physiol 2021; 321:F689-F704. [PMID: 34693742 PMCID: PMC8714974 DOI: 10.1152/ajprenal.00222.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Macula densa (MD) cells, a chief sensory cell type in the nephron, are endowed with unique microanatomic features including a high density of protein synthetic organelles and secretory vesicles in basal cell processes ("maculapodia") that suggest a so far unknown high rate of MD protein synthesis. This study aimed to explore the rate and regulation of MD protein synthesis and their effects on glomerular function using novel transgenic mouse models, newly established fluorescence cell biology techniques, and intravital microscopy. Sox2-tdTomato kidney tissue sections and an O-propargyl puromycin incorporation-based fluorescence imaging assay showed that MD cells have the highest level of protein synthesis within the kidney cortex followed by intercalated cells and podocytes. Genetic gain of function of mammalian target of rapamycin (mTOR) signaling specifically in MD cells (in MD-mTORgof mice) or their physiological activation by low-salt diet resulted in further significant increases in the synthesis of MD proteins. Specifically, these included both classic and recently identified MD-specific proteins such as cyclooxygenase 2, microsomal prostaglandin E2 synthase 1, and pappalysin 2. Intravital imaging of the kidney using multiphoton microscopy showed significant increases in afferent and efferent arteriole and glomerular capillary diameters and blood flow in MD-mTORgof mice coupled with an elevated glomerular filtration rate. The presently identified high rate of MD protein synthesis that is regulated by mTOR signaling is a novel component of the physiological activation and glomerular hemodynamic regulatory functions of MD cells that remains to be fully characterized.NEW & NOTEWORTHY This study discovered the high rate of protein synthesis in macula densa (MD) cells by applying direct imaging techniques with single cell resolution. Physiological activation and mammalian target of rapamycin signaling played important regulatory roles in this process. This new feature is a novel component of the tubuloglomerular cross talk and glomerular hemodynamic regulatory functions of MD cells. Future work is needed to elucidate the nature and (patho)physiological role of the specific proteins synthesized by MD cells.
Collapse
Affiliation(s)
- Urvi Nikhil Shroff
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
| | - Georgina Gyarmati
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
| | - Audrey Izuhara
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
| | - Sachin Deepak
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
- Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
| | - János Peti-Peterdi
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California
| |
Collapse
|
48
|
Mondragón-Huerta CG, Bautista-Pérez R, Baiza-Gutman LA, Escobar-Sánchez ML, Valle-Mondragón LD, Salas-Garrido CG, Castro-Moreno P, Ibarra-Barajas M. Morphology and cyclooxygenase-2 and renin expression in the kidney of young spontaneously hypertensive rats. Vet Pathol 2021; 59:371-384. [PMID: 34841988 DOI: 10.1177/03009858211052663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The kidneys play an important role in blood pressure regulation under normal and pathological conditions. We examined the histological changes and expression patterns of cyclooxygenase-2, renin, and (pro)renin receptor (PRR) in the renal cortex of prehypertensive spontaneously hypertensive rats (SHRs) and Wistar Kyoto rats (WKYs). Moreover, blood pressure and plasma urea, creatinine, angiotensin II, and angiotensin (1-7) levels were measured. The results showed that both strains had similar blood pressure and plasma urea and creatinine levels. The glomerular area, basement membrane thickness, collagen fiber content, and arterial wall thickness were greater in SHRs than in WKYs. By immunohistochemistry, cyclooxygenase-2 was localized in the macula densa and renal tubules of both strains. In SHRs, cyclooxygenase-2 was detected in a larger number of tubules, and the cortical expression of cyclooxygenase-2 was also increased. In both strains, PRR and renin were localized in the tubular epithelium and juxtaglomerular cells, respectively. In SHRs, PRR immunolocalization was increased in the glomerulus. The cortical expression of immature renin was markedly increased in SHRs compared to that in WKYs, while renin was significantly decreased. These changes were associated with higher plasma angiotensin II levels and lower plasma angiotensin (1-7) levels in SHRs. The results indicate that the kidneys of SHRs showed morphological changes and variations in cortical expression patterns of PRR, cyclooxygenase-2, and renin before the development of hypertension.
Collapse
Affiliation(s)
| | - Rocío Bautista-Pérez
- Instituto Nacional de Cardiología, "Dr. Ignacio Chávez," Ciudad de México, México
| | - Luis A Baiza-Gutman
- Universidad Nacional Autónoma de México, Tlalnepantla, Estado de México, México
| | | | | | | | | | | |
Collapse
|
49
|
Song HY, Wei CM, Zhou WX, Hu HF, Wan QJ. Association between admission hemoglobin level and prognosis in patients with type 2 diabetes mellitus. World J Diabetes 2021; 12:1917-1927. [PMID: 34888016 PMCID: PMC8613662 DOI: 10.4239/wjd.v12.i11.1917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/18/2021] [Accepted: 10/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anaemia is common in patients with chronic kidney disease (CKD) and is a major risk factor that contributes to mortality in such patients. Type 2 diabetes mellitus (T2DM) is one of the leading causes of CKD. The association between admission hemoglobin levels and renal damage in patients with T2DM remains unclear.
AIM To evaluate the relationship between admission hemoglobin levels and prognosis in patients with T2DM.
METHODS We performed a retrospective analysis of 265 consecutive patients presenting with T2DM between 2011 and 2015. The composite endpoint was end-stage renal disease or a 50% reduction in the estimated glomerular filtration rate.
RESULTS In multivariable-adjusted Cox proportional hazards models (adjusting for demographic factors, traditional risk factors, lipids), the adjusted hazard ratios (HRs) for the highest and middle tertiles compared to the lowest tertile of hemoglobin were 0.82 (95%CI: 0.11-6.26, P = 0.8457) and 0.28 (95%CI: 0.09-0.85, P = 0.0246), respectively. However, after further adjustment for glycaemia control, hemoglobin was positively related to the risk of the composite endpoint (HR: 1.05, 95%CI: 0.14-8.09, P = 0.9602) when the highest tertile was compared to the lowest tertile of hemoglobin. We found a U-shaped relationship between hemoglobin levels and the composite endpoint. The curve tended to reach the lowest level at an optimal hemoglobin level.
CONCLUSION Among patients with T2DM, a U-shaped relationship was observed between hemoglobin levels and renal damage. A lower admission hemoglobin level (hemoglobin < 13.3 g/dL) is an independent predictor of renal damage.
Collapse
Affiliation(s)
- Hai-Ying Song
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
- Department of Nephrology, Shenzhen University Health Science Center, Shenzhen 518035, Guangdong Province, China
| | - Cui-Mei Wei
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
- Department of Nephrology, Shenzhen University Health Science Center, Shenzhen 518035, Guangdong Province, China
| | - Wen-Xiong Zhou
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
- Department of Nephrology, Shenzhen University Health Science Center, Shenzhen 518035, Guangdong Province, China
| | - Hao-Fei Hu
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
- Department of Nephrology, Shenzhen University Health Science Center, Shenzhen 518035, Guangdong Province, China
| | - Qi-Jun Wan
- Department of Nephrology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, Guangdong Province, China
- Department of Nephrology, Shenzhen University Health Science Center, Shenzhen 518035, Guangdong Province, China
| |
Collapse
|
50
|
Zhang J, Wang X, Cui Y, Jiang S, Wei J, Chan J, Thalakola A, Le T, Xu L, Zhao L, Wang L, Jiang K, Cheng F, Patel T, Buggs J, Vallon V, Liu R. Knockout of Macula Densa Neuronal Nitric Oxide Synthase Increases Blood Pressure in db/db Mice. Hypertension 2021; 78:1760-1770. [PMID: 34657443 DOI: 10.1161/hypertensionaha.121.17643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jie Zhang
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Ximing Wang
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa.,Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China (X.W.)
| | - Yu Cui
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (Y.C., L.Z.)
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Jin Wei
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Jenna Chan
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Anish Thalakola
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Thanh Le
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Lan Xu
- College of Medicine, College of Public Health (L.X.), University of South Florida, Tampa
| | - Liang Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (Y.C., L.Z.)
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| | - Kun Jiang
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center, Research Institute, Tampa, FL (K.J.)
| | - Feng Cheng
- Department of Pharmaceutical Science, College of Pharmacy (F.C.), University of South Florida, Tampa
| | - Trushar Patel
- Department of Urology (T.P.), University of South Florida, Tampa
| | - Jacentha Buggs
- Advanced Organ Disease and Transplantation Institute, Tampa General Hospital, FL (J.B.)
| | - Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA (V.V.)
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology (J.Z., X.W., S.J., J.W., J.C., A.T., T.L., L.W., R.L.), University of South Florida, Tampa
| |
Collapse
|