1
|
Guo ZS, Lu MM, Liu DW, Zhou CY, Liu ZS, Zhang Q. Identification of amino acids metabolomic profiling in human plasma distinguishes lupus nephritis from systemic lupus erythematosus. Amino Acids 2024; 56:56. [PMID: 39292313 PMCID: PMC11410987 DOI: 10.1007/s00726-024-03418-1] [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: 05/26/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Lupus nephritis (LN) is an immunoinflammatory glomerulonephritis associated with renal involvement in systemic lupus erythematosus (SLE). Given the close relationship between plasma amino acids (AAs) and renal function, this study aimed to elucidate the plasma AA profiles in LN patients and identify key AAs and diagnostic patterns that distinguish LN patients from those with SLE and healthy controls. Participants were categorized into three groups: normal controls (NC), SLE, and LN. Ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was employed to quantify AA levels in human plasma. Principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were utilized to identify key AAs. The diagnostic capacity of the models was assessed using receiver operating characteristic (ROC) curve analysis and area under the ROC curve (AUC) values. Significant alterations in plasma AA profiles were observed in LN patients compared to the SLE and NC groups. The OPLS-DA model effectively separated LN patients from the SLE and NC groups. A joint model using histidine (His), lysine (Lys), and tryptophan (Trp) demonstrated exceptional diagnostic performance, achieving an AUC of 1.0 with 100% sensitivity, specificity, and accuracy in predicting LN. Another joint model comprising arginine (Arg), valine (Val), and Trp also exhibited robust predictive performance, with an AUC of 0.998, sensitivity of 93.80%, specificity of 100%, and accuracy of 95.78% in distinguishing between SLE and LN. The joint forecasting models showed excellent predictive capabilities in identifying LN and categorizing lupus disease status. This approach provides a novel perspective for the early identification, prevention, treatment, and management of LN based on variations in plasma AA levels.
Collapse
Affiliation(s)
- Zui-Shuang Guo
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China
| | - Man-Man Lu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China
| | - Chun-Yu Zhou
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China
- Blood Purification Center, First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.
| | - Qing Zhang
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P.R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.
| |
Collapse
|
2
|
Kuypers DRJ, Kamphorst JJ, de Loor H, O'Day EM. Perspective: metabolomics has the potential to change the landscape of kidney transplantation diagnostics. Biomark Med 2024; 18:787-794. [PMID: 39234983 PMCID: PMC11457662 DOI: 10.1080/17520363.2024.2394383] [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: 05/17/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024] Open
Abstract
Kidney transplantation is the most efficient renal replacement therapy. Current diagnostics for monitoring graft health are either invasive or lack precision. Metabolomics is an emerging discipline focused on the analysis of the small molecules involved in metabolism. Given the kidneys' central role in metabolic homeostasis and previous observations of altered metabolites correlating with restricted kidney graft function, metabolomics is highly promising for the discovery of novel biomarkers and the development of novel diagnostics. In this perspective, we summarize the known metabolic roles for the kidney, discuss biomarkers of graft health and immune status emerging from metabolomics research, and provide our perspective on how these and future findings can be integrated in clinical practice to enable precision diagnostics.
Collapse
Affiliation(s)
- Dirk R J Kuypers
- Department of Nephrology & Renal Transplantation, University Hospitals Leuven, Belgium
- Department of Microbiology, Immunology & Transplantation, Nephrology & Renal Transplantation Research Group, KU Leuven, Belgium
| | | | - Henriette de Loor
- Department of Nephrology & Renal Transplantation, University Hospitals Leuven, Belgium
| | | |
Collapse
|
3
|
Semenikhina M, Bohovyk R, Fedoriuk M, Stefanenko M, Klemens CA, Oates JC, Staruschenko A, Palygin O. Renin-angiotensin system-mediated nitric oxide signaling in podocytes. Am J Physiol Renal Physiol 2024; 327:F532-F542. [PMID: 39024356 PMCID: PMC11460333 DOI: 10.1152/ajprenal.00316.2023] [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: 10/09/2023] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
Nitric oxide (NO) is widely recognized for its role in regulating renal function and blood pressure. However, the precise mechanisms by which NO affects renal epithelial cells remain understudied. Our previous research has shown that NO signaling in glomerular podocytes can be initiated by Angiotensin II (ANG II) but not by ATP. This study aims to elucidate the crucial interplay between the renin-angiotensin system (RAS) and NO production in podocytes. To conduct our research, we used cultured human podocytes and freshly isolated rat glomeruli. A variety of RAS peptides were used, alongside confocal microscopy, to detect NO production and NO/Ca2+ cross talk. Dynamic changes in the podocyte cytoskeleton, mediated by RAS-NO intracellular signaling, were observed using fluorescent labeling for F-actin and scanning probe microscopy. The experiments demonstrated that ANG II and ANG III generated high levels of NO by activating the angiotensin II type 2 receptor (AT2R). We did not detect functional MAS receptor presence in podocytes, and the moderate NO response to ANG 1-7 was also mediated through AT2R. Furthermore, NO production impacted intracellular Ca2+ signaling and correlated with an increase in podocyte volume and growth. Scanning probe experiments revealed that AT2R activation and the corresponding NO generation are responsible for the protrusion of podocyte lamellipodia. Taken together, our data indicate that AT2R activation enhances NO production in podocytes and subsequently mediates changes in Ca2+ signaling and podocyte volume dynamics. These mechanisms may play a significant role in both physiological and pathophysiological interactions between the RAS and podocytes.NEW & NOTEWORTHY The renin-angiotensin system plays a crucial role in the production of intracellular nitric oxide within podocytes. This mechanism operates through the activation of the angiotensin II type 2 receptor, leading to dynamic modifications in intracellular calcium levels and the actin filament network. This intricate process is vital for linking the activity of angiotensin receptors to podocyte function.
Collapse
Affiliation(s)
- Marharyta Semenikhina
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Ruslan Bohovyk
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
| | - Mykhailo Fedoriuk
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Mariia Stefanenko
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Christine A Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, United States
| | - Jim C Oates
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, United States
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, United States
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida, United States
- James A. Haley Veterans' Hospital, Tampa, Florida, United States
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
4
|
Demirci H, Popovic S, Dittmayer C, Yilmaz DE, El-Shimy IA, Mülleder M, Hinze C, Su M, Mertins P, Kirchner M, Osmanodja B, Paliege A, Budde K, Amann K, Persson PB, Mutig K, Bachmann S. Immunosuppression with cyclosporine versus tacrolimus shows distinctive nephrotoxicity profiles within renal compartments. Acta Physiol (Oxf) 2024; 240:e14190. [PMID: 38884453 DOI: 10.1111/apha.14190] [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: 04/15/2024] [Revised: 05/18/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
AIM Calcineurin inhibitors (CNIs) are the backbone for immunosuppression after solid organ transplantation. Although successful in preventing kidney transplant rejection, their nephrotoxic side effects contribute to allograft injury. Renal parenchymal lesions occur for cyclosporine A (CsA) as well as for the currently favored tacrolimus (Tac). We aimed to study whether chronic CsA and Tac exposures, before reaching irreversible nephrotoxic damage, affect renal compartments differentially and whether related pathogenic mechanisms can be identified. METHODS CsA and Tac were administered chronically in wild type Wistar rats using osmotic minipumps over 4 weeks. Functional parameters were controlled. Electron microscopy, confocal, and 3D-structured illumination microscopy were used for histopathology. Clinical translatability was tested in human renal biopsies. Standard biochemical, RNA-seq, and proteomic technologies were applied to identify implicated molecular pathways. RESULTS Both drugs caused significant albeit differential damage in vasculature and nephron. The glomerular filtration barrier was more affected by Tac than by CsA, showing prominent deteriorations in endothelium and podocytes along with impaired VEGF/VEGFR2 signaling and podocyte-specific gene expression. By contrast, proximal tubule epithelia were more severely affected by CsA than by Tac, revealing lysosomal dysfunction, enhanced apoptosis, impaired proteostasis and oxidative stress. Lesion characteristics were confirmed in human renal biopsies. CONCLUSION We conclude that pathogenetic alterations in the renal compartments are specific for either treatment. Considering translation to the clinical setting, CNI choice should reflect individual risk factors for renal vasculature and tubular epithelia. As a step in this direction, we share protein signatures identified from multiomics with potential pathognomonic relevance.
Collapse
Affiliation(s)
- Hasan Demirci
- Institute of Functional Anatomy, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Cell- and Neurobiology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Suncica Popovic
- Institute of Functional Anatomy, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Dittmayer
- Department of Neuropathology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Duygu Elif Yilmaz
- Institute of Functional Anatomy, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Ismail Amr El-Shimy
- Molecular Epidemiology Unit, Berlin Institute of Health, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility-High-Throughput Mass Spectrometry, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Hinze
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Mingzhen Su
- Department of Cell- and Neurobiology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité, Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité, Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Paliege
- Department of Nephrology, Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Pontus B Persson
- Department of Translational Physiology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Kerim Mutig
- Department of Translational Physiology, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Sebastian Bachmann
- Institute of Functional Anatomy, Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Cell- and Neurobiology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
5
|
Kamenshchikov NO, Podoksenov YK, Kozlov BN, Maslov LN, Mukhomedzyanov AV, Tyo MA, Boiko AM, Margolis NY, Boshchenko AA, Serebryakova ON, Dzyuman AN, Shirshin AS, Buranov SN, Selemir VD. The Nephroprotective Effect of Nitric Oxide during Extracorporeal Circulation: An Experimental Study. Biomedicines 2024; 12:1298. [PMID: 38927505 PMCID: PMC11201384 DOI: 10.3390/biomedicines12061298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
This study aims to determine the effectiveness of administering 80 ppm nitric oxide in reducing kidney injury, mitochondrial dysfunction and regulated cell death in kidneys during experimental perfusion. Twenty-four sheep were randomized into four groups: two groups received 80 ppm NO conditioning with 90 min of cardiopulmonary bypass (CPB + NO) or 90 min of CPB and hypothermic circulatory arrest (CPB + CA + NO), while two groups received sham protocols (CPB and CPB + CA). Kidney injury was assessed using laboratory (neutrophil gelatinase-associated lipocalin, an acute kidney injury biomarker) and morphological methods (morphometric histological changes in kidney biopsy specimens). A kidney biopsy was performed 60 min after weaning from mechanical perfusion. NO did not increase the concentrations of inhaled NO2 and methemoglobin significantly. The NO-conditioning groups showed less severe kidney injury and mitochondrial dysfunction, with statistical significance in the CPB + NO group and reduced tumor necrosis factor-α expression as a trigger of apoptosis and necroptosis in renal tissue in the CPB + CA + NO group compared to the CPB + CA group. The severity of mitochondrial dysfunction in renal tissue was insignificantly lower in the NO-conditioning groups. We conclude that NO administration is safe and effective at reducing kidney injury, mitochondrial dysfunction and regulated cell death in kidneys during experimental CPB.
Collapse
Affiliation(s)
- Nikolay O. Kamenshchikov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Yuri K. Podoksenov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Boris N. Kozlov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Leonid N. Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Alexander V. Mukhomedzyanov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Mark A. Tyo
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Alexander M. Boiko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Natalya Yu. Margolis
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Alla A. Boshchenko
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111a Kievskaya St., Tomsk 634012, Russia; (Y.K.P.); (B.N.K.); (L.N.M.); (A.V.M.); (M.A.T.); (A.M.B.); (N.Y.M.); (A.A.B.)
| | - Olga N. Serebryakova
- Department of Morphology and General Pathology, Siberian State Medical University, 2 Moskovsky trakt, Tomsk 634050, Russia; (O.N.S.); (A.N.D.)
| | - Anna N. Dzyuman
- Department of Morphology and General Pathology, Siberian State Medical University, 2 Moskovsky trakt, Tomsk 634050, Russia; (O.N.S.); (A.N.D.)
| | - Alexander S. Shirshin
- Federal State Unitary Enterprise “Russian Federal Nuclear Center—All-Russian Research Institute of Experimental Physics”, 37, Mira Ave., Nizhny Novgorod Region, Sarov 607190, Russia; (A.S.S.); (S.N.B.); (V.D.S.)
| | - Sergey N. Buranov
- Federal State Unitary Enterprise “Russian Federal Nuclear Center—All-Russian Research Institute of Experimental Physics”, 37, Mira Ave., Nizhny Novgorod Region, Sarov 607190, Russia; (A.S.S.); (S.N.B.); (V.D.S.)
| | - Victor D. Selemir
- Federal State Unitary Enterprise “Russian Federal Nuclear Center—All-Russian Research Institute of Experimental Physics”, 37, Mira Ave., Nizhny Novgorod Region, Sarov 607190, Russia; (A.S.S.); (S.N.B.); (V.D.S.)
| |
Collapse
|
6
|
Xu C, Tsihlis G, Chau K, Trinh K, Rogers NM, Julovi SM. Novel Perspectives in Chronic Kidney Disease-Specific Cardiovascular Disease. Int J Mol Sci 2024; 25:2658. [PMID: 38473905 PMCID: PMC10931927 DOI: 10.3390/ijms25052658] [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: 01/19/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Chronic kidney disease (CKD) affects > 10% of the global adult population and significantly increases the risk of cardiovascular disease (CVD), which remains the leading cause of death in this population. The development and progression of CVD-compared to the general population-is premature and accelerated, manifesting as coronary artery disease, heart failure, arrhythmias, and sudden cardiac death. CKD and CV disease combine to cause multimorbid cardiorenal syndrome (CRS) due to contributions from shared risk factors, including systolic hypertension, diabetes mellitus, obesity, and dyslipidemia. Additional neurohormonal activation, innate immunity, and inflammation contribute to progressive cardiac and renal deterioration, reflecting the strong bidirectional interaction between these organ systems. A shared molecular pathophysiology-including inflammation, oxidative stress, senescence, and hemodynamic fluctuations characterise all types of CRS. This review highlights the evolving paradigm and recent advances in our understanding of the molecular biology of CRS, outlining the potential for disease-specific therapies and biomarker disease detection.
Collapse
Affiliation(s)
- Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
| | - George Tsihlis
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
| | - Katrina Chau
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
- Blacktown Clinical School, School of Medicine, Western Sydney University, Sydney, NSW 2148, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Department of Renal Services, Blacktown Hospital, Blacktown, NSW 2148, Australia;
| | - Natasha M. Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Renal and Transplantation Medicine, Westmead Hospital, Westmead, NSW 2145, Australia;
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| | - Sohel M. Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW 2145, Australia; (C.X.); (K.T.)
- Faculty of Medicine and Health, The University of Sydney, Science Rd., Camperdown, NSW 2050, Australia
| |
Collapse
|
7
|
Chen Z, Wu S, Huang L, Li J, Li X, Zeng Y, Chen Z, Chen M. Colonic microflora and plasma metabolite-based comparative analysis of unilateral ureteral obstruction-induced chronic kidney disease after treatment with the Chinese medicine FuZhengHuaYuJiangZhuTongLuo and AST-120. Heliyon 2024; 10:e24987. [PMID: 38333870 PMCID: PMC10850519 DOI: 10.1016/j.heliyon.2024.e24987] [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: 10/18/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
Background Many researchers have investigated the use of Chinese herbs to delay the progression of chronic kidney disease (CKD) through their effects on colonic microflora and microbiota-derived metabolites. However, whether FuZhengHuaYuJiangZhuTongLuo (FZHY) has effects that are similar to those of AST-120 on CKD needs to be elucidated. Methods In this study, we compared the effects of FZHY and AST-120 on the colonic microbiota and plasma metabolites in the CKD rat model. We developed a unilateral ureteral obstruction (UUO)-induced CKD rat model and then administered FZHY and AST-120 to these model rats. Non-targeted metabolomic LC-MS analysis, 16S rRNA sequencing, and histopathological staining were performed on plasma, stool, and kidney tissues, respectively, and the joint correlation between biomarkers and metabolites of candidate bacteria was analyzed. Results Our results showed that administering FZHY and AST-120 effectively ameliorated UUO-induced abnormal renal function and renal fibrosis and regulated the composition of microbiota and metabolites. Compared to the UUO model group, the p_Firmicutes and o_Peptostreptococcales_Tissierellales were increased, while 14 negative ion metabolites were upregulated and 21 were downregulated after FZHY treatment. Additionally, 40 positive ion metabolites were upregulated and 63 were downregulated. On the other hand, AST-120 treatment resulted in an increase in the levels of g_Prevotellaceae_NK3B31_group and f_Prevotellaceae, as well as 12 upregulated and 23 downregulated negative ion metabolites and 56 upregulated and 63 downregulated positive ion metabolites. Besides, FZHY increased the levels of candidate bacterial biomarkers that were found to be negatively correlated with some poisonous metabolites, such as 4-hydroxyretinoic acid, and positively correlated with beneficial metabolites, such as l-arginine. AST-120 increased the levels of candidate bacterial biomarkers that were negatively correlated with some toxic metabolites, such as glycoursodeoxycholic acid, 4-ethylphenol, and indole-3-acetic acid. Conclusion FZHY and AST-120 effectively reduced kidney damage, in which, the recovery of some dysregulated bacteria and metabolites are probably involved. As their mechanisms of regulation were different, FZHY might play a complementary role to AST-120 in treating CKD.
Collapse
Affiliation(s)
- Ziwei Chen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Shaobo Wu
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Li Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Jing Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Xueying Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Yu Zeng
- Department of Clinical Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| | - Zejun Chen
- Department of Nephrology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu Traditional Chinese and Western Medicine Hospital, Chengdu First People's Hospital, Chengdu, Sichuan 610072, China
| | - Ming Chen
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, China
| |
Collapse
|
8
|
Ene CD, Tampa M, Georgescu SR, Matei C, Leulescu IMT, Dogaru CI, Penescu MN, Nicolae I. Disturbances in Nitric Oxide Cycle and Related Molecular Pathways in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2023; 15:5797. [PMID: 38136342 PMCID: PMC10741465 DOI: 10.3390/cancers15245797] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/03/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
It is important to note that maintaining adequate levels of nitric oxide (NO), the turnover, and the oxidation level of nitrogen are essential for the optimal progression of cellular processes, and alterations in the NO cycle indicate a crucial step in the onset and progression of multiple diseases. Cellular accumulation of NO and reactive nitrogen species in many types of tumour cells is expressed by an increased susceptibility to oxidative stress in the tumour microenvironment. Clear cell renal cell carcinoma (ccRCC) is a progressive metabolic disease in which tumour cells can adapt to metabolic reprogramming to enhance NO production in the tumour space. Understanding the factors governing NO biosynthesis metabolites in ccRCC represents a relevant, valuable approach to studying NO-based anticancer therapy. Exploring the molecular processes mediated by NO, related disturbances in molecular pathways, and NO-mediated signalling pathways in ccRCC could have significant therapeutic implications in managing and treating this condition.
Collapse
Affiliation(s)
- Corina Daniela Ene
- Department of Nephrology, Carol Davila Clinical Hospital of Nephrology, 010731 Bucharest, Romania; (C.D.E.); (M.N.P.)
- Department of Nephrology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mircea Tampa
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania; (I.M.T.L.); (C.I.D.); (I.N.)
| | - Simona Roxana Georgescu
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania; (I.M.T.L.); (C.I.D.); (I.N.)
| | - Clara Matei
- Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Iulia Maria Teodora Leulescu
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania; (I.M.T.L.); (C.I.D.); (I.N.)
| | - Claudia Ioana Dogaru
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania; (I.M.T.L.); (C.I.D.); (I.N.)
| | - Mircea Nicolae Penescu
- Department of Nephrology, Carol Davila Clinical Hospital of Nephrology, 010731 Bucharest, Romania; (C.D.E.); (M.N.P.)
- Department of Nephrology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ilinca Nicolae
- Department of Dermatology, “Victor Babes” Clinical Hospital for Infectious Diseases, 030303 Bucharest, Romania; (I.M.T.L.); (C.I.D.); (I.N.)
| |
Collapse
|
9
|
Mitsnefes MM, Wühl E. Role of hypertension in progression of pediatric CKD. Pediatr Nephrol 2023; 38:3519-3528. [PMID: 36732375 DOI: 10.1007/s00467-023-05894-1] [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: 12/13/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 02/04/2023]
Abstract
Hypertension is frequent in children with chronic kidney disease (CKD). Its prevalence varies according to CKD stage and cause. It is relatively uncommon in children with congenital kidney disease, while acquired kidney disease is associated with a higher prevalence of hypertension. Studies in children with CKD utilizing ambulatory blood pressure monitoring also showed a high prevalence of masked hypertension. Uncontrolled and longstanding hypertension in children is associated with progression of CKD. Aggressive treatment of high blood pressure should be an essential part of care to delay CKD progression in children.
Collapse
Affiliation(s)
- Mark M Mitsnefes
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229, USA.
| | - Elke Wühl
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
10
|
Koppara NK, Medooru KK, Yadagiri LA, Vishnubotla SK, Rapur R, Bitla AR. A Study of Oxidative Stress, Inflammation, and Endothelial Dysfunction in Diabetic and Nondiabetic Chronic Kidney Disease Pre-Dialysis Patients. Indian J Nephrol 2023; 33:420-425. [PMID: 38174313 PMCID: PMC10752391 DOI: 10.4103/ijn.ijn_222_22] [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: 06/26/2022] [Revised: 10/13/2022] [Accepted: 11/29/2022] [Indexed: 01/05/2024] Open
Abstract
Background Oxidative stress, inflammation, and endothelial dysfunction represent a key triad for the development and progression of atherosclerosis. Due to chronic low-grade inflammation in chronic kidney disease (CKD), concentrations of various inflammatory, endothelial, and oxidative stress markers are elevated, increasing the risk of atherosclerosis. The present study was undertaken to compare oxidative stress, inflammation, and endothelial dysfunction in diabetic and nondiabetic CKD pre-dialysis patients. Materials and Methods This was an observational study on 120 CKD pre-dialysis patients: 60 with diabetes and 60 without diabetes. Markers of oxidative stress were measured in blood - malondialdehyde (MDA), ferric reducing ability of plasma (FRAP), paroxonase-1 (PON-1), ischemia-modified albumin (IMA); inflammation - interleukin-6 (IL-6), high-sensitivity C-reactive protein (hsCRP); and endothelial dysfunction - nitric oxide (NO), carotid wall intima-media thickness (CIMT). Comparisons between the two groups for continuous variables were made with the Student's unpaired t-test or Mann-Whitney test and for categorical values with χ2-test, as appropriate. Results MDA, IMA, IL-6, hsCRP, NO, and CIMT were significantly higher, while FRAP and PON-1 were significantly lower in the diabetic group when compared to nondiabetic group (P < 0.001). The number of atherosclerotic plaques was also significantly higher in the diabetic group compared to nondiabetic group. Conclusion Our study showed increased oxidative stress, inflammation, endothelial dysfunction, and atherosclerosis in diabetic CKD pre-dialysis patients when compared to nondiabetic CKD pre-dialysis patients and in late stages when compared to early stages of CKD in both groups, indicating increased cardiovascular risk in late stages and diabetic CKD pre-dialysis patients.
Collapse
Affiliation(s)
- Naveen Kumar Koppara
- Department of Nephrology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Kusuma Kumari Medooru
- Department of Biochemistry, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | | | - Siva Kumar Vishnubotla
- Department of Nephrology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Ram Rapur
- Department of Nephrology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Aparna R. Bitla
- Department of Biochemistry, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| |
Collapse
|
11
|
Forsse JS, Richardson KA, Torres R, Lowry C, Taylor JK, Beeson CL, Ward J, Dhillon A, Niceler B, Ismaeel A, Koutakis P. Exploring an Unknown Corner of a Well-Known Topic: HIIE Influence on Renal Health and Filtration in Healthy Individuals Free of Cardiometabolic Diseases. Sports (Basel) 2023; 11:210. [PMID: 37999427 PMCID: PMC10675315 DOI: 10.3390/sports11110210] [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: 08/27/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Aerobic exercise, specifically high-intensity interval exercise (HIIE), and its effects on renal health and filtration (RHF) are not well understood. Several studies support incorporating contemporary biomarkers serum cystatin C (CyC) and urine epidermal growth factor (uEGF) to combat the volatility of serum creatinine (sCr). Using these biomarkers, we examined the acute influences HIIE has on RHF to determine if there is a ceiling effect in healthy populations. The purpose was to determine the influence of an acute bout of HIIE on RHF. Thirty-six participants (n = 22 males; n = 14 females; age 37.6 ± 12.4 years.; BF% 19.2 ± 7.1%; VO2max 41.8 + 7.4 mL/kg/min) completed 30 min of HIIE on a treadmill (80% and 40% of VO2reserve in 3:2 min ratio). Blood and urine samples were obtained under standardized conditions before, 1 h, and 24 h post-exercise. CyC, sCR, uEGF, urine creatinine (uCr), uCr/uEGF ratio, and multiple estimates of glomerular filtration rate (eGFR) Modification of Diet in Renal Disease (MDRD) and CKD-EPI equations were used. The analysis employed paired sample t-tests and repeated measures ANOVAs. CyC, uEGF, uCr, and uCr/uEGF ratio concentrations were not altered between timepoints. sCr increased 1 h post-exercise (p > 0.002) but not at 24 h post-exercise. eGFR decreased in the MDRD and CKD-EPI equations at 1 h (p > 0.012) with no changes at 24 h post-exercise. CyC and sCr/CyC demonstrated no significant changes. CyC and uEGF are not altered by acute HIIE. The results demonstrate a potential ceiling effect in contemporary and traditional biomarkers of RHF, indicating improvements in RHF may be isolated to populations with reduced kidney function.
Collapse
Affiliation(s)
- Jeffrey S. Forsse
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Kathleen A. Richardson
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Ricardo Torres
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Catherine Lowry
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO 80011, USA
| | - James Kyle Taylor
- Medical & Clinical Laboratory Sciences, Auburn University—Montgomery, Montgomery, AL 36124, USA;
| | - Cassidy L. Beeson
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
| | - Jacob Ward
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Southern Illinois University Medical School, Lindegren Hall, 600 Agriculture Dr #132, Carbondale, IL 62901, USA
| | - Anurag Dhillon
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Health Science Center, San Antonio Joe R and Teresa Lozano Long School of Medicine, The University of Texas, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Brock Niceler
- Integrated Laboratory of Exercise, Nutrition, and Renal Vascular Research, Department of Health, Human Performance, and Recreation, Baylor University, Waco, TX 76706, USA; (K.A.R.); (R.T.); (C.L.); (C.L.B.); (J.W.); (A.D.); (B.N.)
- Waco Family Medicine, Waco, TX 76707, USA
| | - Ahmed Ismaeel
- Department of Physiology, University of Kentucky, 780 Rose Street, MS508, Lexington, KY 40536, USA;
- Clinical Muscle Biology Lab, Baylor University, Waco, TX 76706, USA;
| | | |
Collapse
|
12
|
Liu H, Huang Y, Huang M, Wang M, Ming Y, Chen W, Chen Y, Tang Z, Jia B. From nitrate to NO: potential effects of nitrate-reducing bacteria on systemic health and disease. Eur J Med Res 2023; 28:425. [PMID: 37821966 PMCID: PMC10566198 DOI: 10.1186/s40001-023-01413-y] [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: 01/19/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
Current research has described improving multisystem disease and organ function through dietary nitrate (DN) supplementation. They have provided some evidence that these floras with nitrate (NO3-) reductase are mediators of the underlying mechanism. Symbiotic bacteria with nitrate reductase activity (NRA) are found in the human digestive tract, including the mouth, esophagus and gastrointestinal tract (GT). Nitrate in food can be converted to nitrite under the tongue or in the stomach by these symbiotic bacteria. Then, nitrite is transformed to nitric oxide (NO) by non-enzymatic synthesis. NO is currently recognized as a potent bioactive agent with biological activities, such as vasodilation, regulation of cardiomyocyte function, neurotransmission, suppression of platelet agglutination, and prevention of vascular smooth muscle cell proliferation. NO also can be produced through the conventional L-arginine-NO synthase (L-NOS) pathway, whereas endogenous NO production by L-arginine is inhibited under hypoxia-ischemia or disease conditions. In contrast, exogenous NO3-/NO2-/NO activity is enhanced and becomes a practical supplemental pathway for NO in the body, playing an essential role in various physiological activities. Moreover, many diseases (such as metabolic or geriatric diseases) are primarily associated with disorders of endogenous NO synthesis, and NO generation from the exogenous NO3-/NO2-/NO route can partially alleviate the disease progression. The imbalance of NO in the body may be one of the potential mechanisms of disease development. Therefore, the impact of these floras with nitrate reductase on host systemic health through exogenous NO3-/NO2-/NO pathway production of NO or direct regulation of floras ecological balance is essential (e.g., regulation of body homeostasis, amelioration of diseases, etc.). This review summarizes the bacteria with nitrate reductase in humans, emphasizing the relationship between the metabolic processes of this microflora and host systemic health and disease. The potential effects of nitrate reduction bacteria on human health and disease were also highlighted in disease models from different human systems, including digestive, cardiovascular, endocrine, nervous, respiratory, and urinary systems, providing innovative ideas for future disease diagnosis and treatment based on nitrate reduction bacteria.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yisheng Huang
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Mingshu Huang
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Min Wang
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yue Ming
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weixing Chen
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yuanxin Chen
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Zhengming Tang
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Bo Jia
- Department of Oral Surgery, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
13
|
Sinha F, Schweda F, Maier LS, Wagner S. Impact of Impaired Kidney Function on Arrhythmia-Promoting Cardiac Ion Channel Regulation. Int J Mol Sci 2023; 24:14198. [PMID: 37762501 PMCID: PMC10532292 DOI: 10.3390/ijms241814198] [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: 08/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is associated with a significantly increased risk of cardiovascular events and sudden cardiac death. Although arrhythmias are one of the most common causes of sudden cardiac death in CKD patients, the molecular mechanisms involved in the development of arrhythmias are still poorly understood. In this narrative review, therefore, we summarize the current knowledge on the regulation of cardiac ion channels that contribute to arrhythmia in CKD. We do this by first explaining the excitation-contraction coupling, outlining current translational research approaches, then explaining the main characteristics in CKD patients, such as abnormalities in electrolytes and pH, activation of the autonomic nervous system, and the renin-angiotensin-aldosterone system, as well as current evidence for proarrhythmic properties of uremic toxins. Finally, we discuss the substance class of sodium-glucose co-transporter 2 inhibitors (SGLT2i) on their potential to modify cardiac channel regulation in CKD and, therefore, as a treatment option for arrhythmias.
Collapse
Affiliation(s)
- Frederick Sinha
- Department for Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany; (F.S.)
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, 93053 Regensburg, Germany
| | - Lars S. Maier
- Department for Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany; (F.S.)
| | - Stefan Wagner
- Department for Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany; (F.S.)
| |
Collapse
|
14
|
Tain YL, Hsu CN. The NOS/NO System in Renal Programming and Reprogramming. Antioxidants (Basel) 2023; 12:1629. [PMID: 37627624 PMCID: PMC10451971 DOI: 10.3390/antiox12081629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Nitric oxide (NO) is a gaseous signaling molecule with renoprotective properties. NO can be produced in NO synthase (NOS)-dependent or -independent manners. NO deficiency plays a decisive role in chronic kidney disease (CKD). Kidney development can be affected in response to adverse intrauterine conditions that induce renal programming, thereby raising the risk of developing CKD in adulthood. Conversely, detrimental programming processes could be postponed or halted prior to the onset of CKD by early treatments, namely reprogramming. The current review provides an overview of the NOS/NO research performed in the context of renal programming and reprogramming. NO deficiency has been increasingly found to interact with the different mechanisms behind renal programming, such as oxidative stress, aberrant function of the renin-angiotensin system, disturbed nutrient-sensing mechanisms, dysregulated hydrogen sulfide signaling, and gut microbiota dysbiosis. The supplementation of NOS substrates, the inhibition of asymmetric dimethylarginine (ADMA), the administration of NO donors, and the enhancement of NOS during gestation and lactation have shown beneficial effects against renal programming in preclinical studies. Although human data on maternal NO deficiency and offspring kidney disease are scarce, experimental data indicate that targeting NO could be a promising reprogramming strategy in the setting of renal programming.
Collapse
Affiliation(s)
- You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
15
|
Amin SN, El-Gamal EM, Rashed LA, Kamar SS, Haroun MA. Inhibition of notch signalling and mesangial expansion by combined glucagon like peptide-1 agonist and crocin therapy in animal model of diabetic nephropathy. Arch Physiol Biochem 2023; 129:544-554. [PMID: 33280420 DOI: 10.1080/13813455.2020.1846203] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is one of the devastating complications in diabetes mellitus (DM). Glucagon-like peptide-1 (GLP-1) is one of the incretins secreted from L cells in the intestine. Crocin (a carotenoid component of saffron) has antioxidants properties. We investigated the renal effects of Exendin-4 as a GLP-1 agonist and Crocin in DN.Thirty male rats were divided into five groups: control, type II DM, type II DM + Exendin-4, type II DM + Crocin and type II DM + Exendine-4 + Crocin. At the end of the experimental period, systolic and diastolic blood pressures were measured, and GFR was calculated. Blood and urine samples were collected for biochemical analysis. Tissue samples were collected from the kidney for histological examination and biochemical measurements of protein expression.Treatment with GLP-1 agonist or Crocin caused a significant improvement in renal function. Better results were achieved with simultaneous administration of both drugs with inhibition of notch signalling pathway and the related proteins.
Collapse
Affiliation(s)
- Shaimaa Nasr Amin
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqaa, Jordan
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman Mumtaz El-Gamal
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samaa Samir Kamar
- Department of Histology and Cell Biology, Cairo University, Cairo, Egypt
| | - Maged Ahmed Haroun
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
16
|
Barman Z, Hasan M, Miah R, Mou AD, Hafsa JM, Trisha AD, Mahmud F, Ali N. Association between hyperuricemia and chronic kidney disease: a cross-sectional study in Bangladeshi adults. BMC Endocr Disord 2023; 23:45. [PMID: 36803682 PMCID: PMC9942427 DOI: 10.1186/s12902-023-01304-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND AND AIMS Chronic kidney disease (CKD) is a public health concern worldwide and has been recognized as a significant risk factor for cardiovascular disease. The elevated level of uric acid (hyperuricemia) has been suggested to be associated with obesity, hypertension, cardiovascular disease and diabetes. However, there is limited information on the relationship between hyperuricemia and CKD. Therefore, this study aimed to estimate the prevalence of CKD and assess its relationship with hyperuricemia in Bangladeshi adults. METHODS In this study, blood samples were collected from 545 participants (398 males and 147 females) aged ≥ 18 years. Biochemical parameters such as serum uric acid (SUA), lipid profile markers, glucose, creatinine and urea were measured by colorimetric methods. The estimated glomerular filtration rate (eGFR) and CKD were determined based on serum creatinine levels with existed formula. Multivariate logistic regression analysis was performed to evaluate the association between SUA and CKD. RESULTS The overall prevalence of CKD was 5.9% with 6.1% in males and 5.2% in females. Hyperuricemia was prevalent in 18.7% of participants with 23.2% in males and 14.6% in females. An increasing trend of CKD prevalence was observed with increasing age in the groups. The mean eGFR level was significantly lower in male (95.1 ± 31.8 ml/min/1.73m2) than in female (109.3 ± 77.4 ml/min/1.73m2) subjects (p < 0.01). The mean level of SUA was significantly higher (p < 0.01) in participants having CKD (7.1 ± 1.9 mg/dL) than in participants without CKD (5.7 ± 1.6 mg/dL). A decreasing trend for eGFR concentration and an increasing trend for CKD prevalence was observed across the SUA quartiles (p < 0.001). In regression analysis, a significant positive association was found between hyperuricemia and CKD. CONCLUSION This study showed an independent association between hyperuricemia and CKD in Bangladeshi adults. Further mechanistic studies are needed to explore the potential link between hyperuricemia and CKD.
Collapse
Affiliation(s)
- Zitu Barman
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Mahmudul Hasan
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Rakib Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Ananya Dutta Mou
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Jaasia Momtahena Hafsa
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Aporajita Das Trisha
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Firoz Mahmud
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh
| | - Nurshad Ali
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, 3114, Bangladesh.
| |
Collapse
|
17
|
Köksal MM, Şekerler T, Çevik Ö, Şener A. Paricalcitol protects against hydrogen peroxide-induced injury in endothelial cells through suppression of apoptosis. Exp Biol Med (Maywood) 2023; 248:186-192. [PMID: 36373746 PMCID: PMC10041050 DOI: 10.1177/15353702221101615] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The vascular endothelium is one of the main targets of oxidative stress which plays an important role in the pathophysiology of vascular damage. Recent studies show that vitamin D can positively regulate endothelial functions in various chronic diseases and in cases of increased oxidative stress. In our study, we investigated the restorative and protective potentials of paricalcitol which is frequently used in patients with chronic renal failure, a vitamin D analogue, in human umbilical vein endothelial cells (HUVEC) before and after H2O2-induced oxidative stress. Paricalcitol treatment after the oxidative stress induced by H2O2 increased cell viability in endothelial cells depending on the dose that was used. While paricalcitol (500 nM) decreased caspase-3 activity and mitochondrial membrane potential loss, it increased nitric oxide (NO) production and reduced glutathione (GSH) levels. Paricalcitol treatment before oxidative stress increased cell viability. It increased NO production and mitochondrial membrane potential while significantly reducing caspase-3 activity. While paricalcitol caused a significant inhibition of protein disulfide isomerase (PDI) reductase activity in healthy endothelial cells, it did not cause a significant change on the PDI reductase activity under oxidative stress conditions. Present study showed that paricalcitol has restorative and protective effects on endothelial cells against oxidative injury, but these effects occur at high concentrations of paricalcitol.
Collapse
Affiliation(s)
- Muhammet Murat Köksal
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, İstanbul 34854, Turkey
| | - Turgut Şekerler
- Department of Biochemistry, Faculty of Pharmacy, Marmara University, İstanbul 34854, Turkey
| | - Özge Çevik
- Department of Medicinal Biochemistry, School of Medicine, Aydın Adnan Menderes University, Aydın 09000, Turkey
| | - Azize Şener
- Department of Biochemistry, Faculty of Pharmacy, Fenerbahçe University, İstanbul 34758, Turkey
| |
Collapse
|
18
|
Nitric-oxide precursors and dimethylarginines as risk markers for accelerated measured GFR decline in the general population. Kidney Int Rep 2023; 8:818-826. [PMID: 37069987 PMCID: PMC10105054 DOI: 10.1016/j.ekir.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/02/2022] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Introduction Nitric oxide (NO) deficiency is associated with endothelial dysfunction, hypertension, atherosclerosis, and chronic kidney disease (CKD). Reduced NO bioavailability is hypothesized to play a vital role in kidney function impairment and CKD. We investigated the association of serum levels of endogenous inhibitors of NO, asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA), and precursors of NO, arginine, citrulline, and ornithine, with a decline in glomerular filtration rate (GFR) and new-onset CKD. Methods In a prospective cohort study of 1407 healthy, middle-aged participants of Northern European origin in the Renal Iohexol Clearance Survey (RENIS), GFR was measured repeatedly with iohexol clearance during a median follow-up time of 11 years. GFR decline rates were analyzed using a linear mixed model, new-onset CKD (GFR < 60 ml/min per 1.73 m2) was analyzed with interval-censored Cox regression, and accelerated GFR decline (the 10% with the steepest GFR decline) was analyzed with logistic regression. Results Higher SDMA was associated with slower annual GFR decline. Higher levels of citrulline and ornithine were associated with accelerated GFR decline (odds ratio [OR], 1.43; 95% confidence interval [CI] 1.16-1.76 per SD higher citrulline and OR 1.23; 95% CI 1.01 to 1.49 per SD higher ornithine). Higher citrulline was associated with new-onset CKD, with a hazard ratio of 1.33 (95% CI 1.07-1.66) per SD higher citrulline. Conclusions Associations between NO precursors and the outcomes suggest that NO metabolism plays a significant role in the pathogenesis of age-related GFR decline and the development of CKD in middle-aged people.
Collapse
|
19
|
Oh ES, Freeberg KA, Steele CN, Wang W, Farmer‐Bailey H, Coppock ME, Seals DR, Chonchol M, Rossman MJ, Craighead DH, Nowak KL. Cerebrovascular pulsatility index is higher in chronic kidney disease. Physiol Rep 2023; 11:e15561. [PMID: 36636757 PMCID: PMC9837424 DOI: 10.14814/phy2.15561] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are more likely to die of cardiovascular diseases, including cerebrovascular disease, than to progress to end-stage kidney disease. Cerebrovascular dysfunction, characterized by reduced cerebrovascular reactivity, cerebral hypoperfusion, and increased pulsatile flow within the brain, precedes the onset of dementia and is linked to cognitive dysfunction. However, whether impaired cerebrovascular function is present in non-dialysis dependent CKD is largely unknown. Using transcranial Doppler, we compared middle cerebral artery (MCA) blood velocity response to hypercapnia (normalized for blood pressure and end-tidal CO2 ; a measure of cerebrovascular reactivity) and MCA pulsatility index (PI; a measure of cerebrovascular stiffness) in patients with stage 3-4 CKD vs. age-matched healthy controls. We also administered the NIH cognitive toolbox (cognitive function), measured carotid-femoral pulse-wave velocity (PWV; aortic stiffness), and assessed ex vivo nitric oxide (NO) and reactive oxygen species (ROS) production from human brain endothelial cells incubated with serum obtained from study participants. MCA PI was higher in patients with CKD vs. controls; however, normalized MCA blood velocity response to hypercapnia did not differ between groups. Similar results were observed in a validation cohort of midlife and older adults divided by the median estimated glomerular filtration rate (eGFR). MCA PI was associated with greater large-elastic artery stiffness (carotid-femoral PWV), worse executive function (trails B time), lower eGFR, and higher ex vivo ROS production. These data suggest that impaired kidney function is associated with greater cerebrovascular stiffness, which may contribute to the known increased risk for cognitive impairment in patients with CKD.
Collapse
Affiliation(s)
- Ester S. Oh
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Kaitlin A. Freeberg
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Cortney N. Steele
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Wei Wang
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Heather Farmer‐Bailey
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - McKinley E. Coppock
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Douglas R. Seals
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Michel Chonchol
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Matthew J. Rossman
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Daniel H. Craighead
- Department of Integrative PhysiologyUniversity of Colorado BoulderBoulderColoradoUSA
| | - Kristen L. Nowak
- Division of Renal Diseases and HypertensionUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
20
|
Effects of Chronic Kidney Disease on Nanomechanics of the Endothelial Glycocalyx Are Mediated by the Mineralocorticoid Receptor. Int J Mol Sci 2022; 23:ijms231810659. [PMID: 36142571 PMCID: PMC9503126 DOI: 10.3390/ijms231810659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 02/03/2023] Open
Abstract
Endothelial mechanics control vascular reactivity and are regulated by the mineralocorticoid receptor (MR) and its downstream target, the epithelial Na+ channel (ENaC). Endothelial dysfunction is a hallmark of chronic kidney disease (CKD), but its mechanisms are poorly understood. We hypothesized that CKD disrupts endothelial mechanics in an MR/ENaC-dependent process. METHODS Primary human endothelial cells were cultured with uremic serum derived from children with stage 3-5 (predialysis) CKD or adult hemodialysis (HD) patients or healthy controls. The height and stiffness of the endothelial glycocalyx (eGC) and cortex were monitored by atomic force microscopy (AFM) using an ultrasensitive mechanical nanosensor. RESULTS In a stage-dependent manner, sera from children with CKD induced a significant increase in eGC and cortex stiffness and an incremental reduction of the eGC height. AFM measurements were significantly associated with individual pulse wave velocity and serum concentrations of gut-derived uremic toxins. Serum from HD patients increased MR expression and mechanical stiffness of the endothelial cortex, an effect reversed by MR and ENaC antagonists, decreased eNOS expression and NO bioavailability, and augmented monocyte adhesion. CONCLUSION These data indicate progressive structural damage of the endothelial surface with diminishing kidney function and identify the MR as a mediator of CKD-induced endothelial dysfunction.
Collapse
|
21
|
Feuer DS, Handberg EM, Mehrad B, Wei J, Bairey Merz CN, Pepine CJ, Keeley EC. Microvascular Dysfunction as a Systemic Disease: A Review of the Evidence. Am J Med 2022; 135:1059-1068. [PMID: 35472396 PMCID: PMC9427712 DOI: 10.1016/j.amjmed.2022.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 12/11/2022]
Abstract
Microvascular dysfunction describes a varied set of conditions that includes vessel destruction, abnormal vasoreactivity, in situ thrombosis, and fibrosis, which ultimately results in tissue damage and progressive organ failure. Microvascular dysfunction has a wide array of clinical presentations, ranging from ischemic heart disease to renal failure, stroke, blindness, pulmonary arterial hypertension, and dementia. An intriguing unifying hypothesis suggests that microvascular dysfunction of specific organs is an expression of a systemic illness that worsens with age and is accelerated by vascular risk factors. Studying relationships across a spectrum of microvascular diseases affecting the brain, retina, kidney, lung, and heart may uncover shared pathologic mechanisms that could inform novel treatment strategies. We review the evidence that supports the notion that microvascular dysfunction represents a global pathologic process. Our focus is on studies reporting concomitant microvascular dysfunction of the heart with that of the brain, kidney, retina, and lung.
Collapse
Affiliation(s)
| | | | - Borna Mehrad
- Department of Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville
| | - Janet Wei
- Barbra Streisand Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, Calif
| | - C Noel Bairey Merz
- Barbra Streisand Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, Calif
| | - Carl J Pepine
- Department of Medicine; Division of Cardiovascular Medicine
| | - Ellen C Keeley
- Department of Medicine; Division of Cardiovascular Medicine.
| |
Collapse
|
22
|
Zeng L, Chen M, Ahmad H, Zheng X, Ouyang Y, Yang P, Yang Z, Gao D, Tian Z. Momordica charantia Extract Confers Protection Against Hypertension in Dahl Salt-Sensitive Rats. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:373-382. [PMID: 35705768 DOI: 10.1007/s11130-022-00971-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
Hypertension is one of the main factors of cardiovascular disease worldwide and is strongly related to the overall mortality. High salt intake is a major risk factors for hypertension. Identifying functional foods that can help prevent mechanistic abnormalities mediating salt-induced hypertension is an issue of considerable nutraceutical and scientific interest. Dietary Momordica charantia may be an alternative approach to avoid salt-induced hypertension. Dahl salt-sensitive (DSS) rats were used to determine whether Momordica charantia water extracts (ME) exerts anti-hypertensive effects in the present study. ME gavage could significantly prevented the increase of blood pressure, blood urea nitrogen, creatinine, and urine protein-to-creatinine ratio of DSS rats. Metabolomics analysis indicated that high-salt diet induced abnormal amino acid metabolism was related to nitric oxide (NO) deficiency, but ME gavage could upregulate the activities of nitric oxide synthase, aspartate aminotransferase, argininosuccinate lyase, argininosuccinate synthase and restore endogenous synthesis of arginine and NO. Meanwhile, renal function was improved after ME gavage. Citrulline, as one of the important component in ME, could attenuate salt-induced hypertension by increasing endogenous synthesis of arginine and NO. Antioxidants in ME, such as phenolic compound, may avoid high-salt induced oxidative stress in DSS rats, which may be another mechanism by which ME prevented blood pressure increase. Thus, the present study indicated that feeding Momordica charantia could avoid high-salt-induced hypertension in DSS rats.
Collapse
Affiliation(s)
- Li Zeng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hussain Ahmad
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xuewei Zheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanan Ouyang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Pengfei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
23
|
Vanent KN, Leasure AC, Acosta JN, Kuohn LR, Woo D, Murthy SB, Kamel H, Messé SR, Mullen MT, Cohen JB, Cohen DL, Townsend RR, Petersen NH, Sansing LH, Gill TM, Sheth KN, Falcone GJ. Association of Chronic Kidney Disease With Risk of Intracerebral Hemorrhage. JAMA Neurol 2022; 79:911-918. [PMID: 35969388 PMCID: PMC9379821 DOI: 10.1001/jamaneurol.2022.2299] [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: 03/24/2022] [Accepted: 06/24/2022] [Indexed: 11/14/2022]
Abstract
Importance The evidence linking chronic kidney disease (CKD) to spontaneous intracerebral hemorrhage (ICH) is inconclusive owing to possible confounding by comorbidities that frequently coexist in patients with these 2 diseases. Objective To determine whether there is an association between CKD and ICH risk. Design, Setting, and Participants A 3-stage study that combined observational and genetic analyses was conducted. First, the association between CKD and ICH risk was tested in the Ethnic/Racial Variations of Intracerebral Hemorrhage (ERICH) study, a multicenter case-control study in the US. All participants with available data on CKD from ERICH were included. Second, this analysis was replicated in the UK Biobank (UKB), an ongoing population study in the UK. All participants in the UKB were included in this study. Third, mendelian randomization analyses were implemented in the UKB using 27 CKD-related genetic variants to test for genetic associations. ERICH was conducted from August 1, 2010, to August 1, 2017, and observed participants for 1 year. The UKB enrolled participants between 2006 and 2010 and will continue to observe them for 30 years. Data analysis was performed from November 11, 2019, to May 10, 2022. Exposures CKD stages 1 to 5. Main Outcomes and Measures The outcome of interest was ICH, ascertained in ERICH via expert review of neuroimages and in the UKB via a combination of self-reported data and International Statistical Classification of Diseases, Tenth Revision, codes. Results In the ERICH study, a total of 2914 participants with ICH and 2954 controls who had available data on CKD were evaluated (mean [SD] age, 61.6 [14.0] years; 2433 female participants [41.5%]; 3435 male participants [58.5%]); CKD was found to be independently associated with higher risk of ICH (odds ratio [OR], 1.95; 95% CI, 1.35-2.89; P < .001). This association was not modified by race and ethnicity. Replication in the UKB with 1341 participants with ICH and 501 195 controls (mean [SD] age, 56.5 [8.1] years; 273 402 female participants [54.4%]; 229 134 male participants [45.6%]) confirmed this association (OR, 1.28; 95% CI, 1.01-1.62; P = .04). Mendelian randomization analyses indicated that genetically determined CKD was associated with ICH risk (OR, 1.56; 95% CI, 1.13-2.16; P = .007). Conclusions and Relevance In this 3-stage study that combined observational and genetic analyses among study participants enrolled in 2 large observational studies with different characteristics and study designs, CKD was consistently associated with higher risk of ICH. Mendelian randomization analyses suggest that this association was causal. Further studies are needed to identify the specific biological pathways that mediate this association.
Collapse
Affiliation(s)
- Kevin N. Vanent
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Audrey C. Leasure
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Julian N. Acosta
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lindsey R. Kuohn
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Santosh B. Murthy
- Clinical and Translational Neuroscience Unit, Department of Neurology, Weill Cornell Medicine, New York, New York
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Department of Neurology, Weill Cornell Medicine, New York, New York
- Deputy Editor, JAMA Neurology
| | - Steven R. Messé
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Michael T. Mullen
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jordana B. Cohen
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Information, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Debbie L. Cohen
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Raymond R. Townsend
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nils H. Petersen
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lauren H. Sansing
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Thomas M. Gill
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kevin N. Sheth
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Guido J. Falcone
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Cofer LB, Soomro QH, Xia Y, Luttrell-Williams E, Myndzar K, Charytan DM, Berger JS. Platelet Activity and Cardiovascular Risk in CKD and Peripheral Artery Disease. Kidney Int Rep 2022; 7:2242-2250. [PMID: 36217517 PMCID: PMC9546761 DOI: 10.1016/j.ekir.2022.07.169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 01/25/2023] Open
Abstract
Introduction Platelet dysfunction and cardiovascular risk are well-recognized features of chronic kidney disease (CKD). Platelets drive the development and progression of cardiovascular disease (CVD). The relationships between kidney function, platelet activity, and cardiovascular risk are poorly defined. Methods We compared platelet activity and incident cardiovascular events by CKD status (estimated glomerular filtration rate [eGFR] < 60 ml/min per 1.73 m2) using data from the Platelet Activity and Cardiovascular Events study, a prospective cohort study that enrolled adults with peripheral artery disease (PAD) undergoing lower extremity revascularization. Platelet activity was measured using light transmission aggregometry (LTA) in response to submaximal dose agonist stimulation, and the subjects were followed for incident adverse cardiovascular events for a median of 18 months. Results Overall, 113 of 285 (40%) subjects had CKD. Subjects with, versus without, CKD had higher platelet aggregation in response to stimulation with adenosine diphosphate (ADP), serotonin, epinephrine, and arachidonic acid (AA) + ex vivo aspirin (P < 0.05 for each). Following multivariable adjustment, subjects with CKD had elevated risk for myocardial infarction (MI) (adjusted hazard ratio 2.2, 95% confidence interval [1.02–4.9]) and major adverse cardiovascular events (MACE) (1.9 [1.2–3.3]) compared to those without CKD. Platelet aggregation in response to submaximal dose agonist stimulation mediated 7% to 26% of the excess risk for cardiovascular events associated with CKD. Conclusion Among subjects with PAD undergoing lower extremity revascularization, CKD is associated with increased platelet activity that mediates, in part, elevated cardiovascular risk.
Collapse
|
25
|
Maassen H, Said MY, Frenay ARS, Koning A, Post A, Riphagen IJ, Heiner-Fokkema MR, Drabert K, Fernandez BO, Gans ROB, van den Berg E, Navis G, Tsikas D, Feelisch M, Bakker SJL, van Goor H. Nitric oxide and long-term outcomes after kidney transplantation: Results of the TransplantLines cohort study. Nitric Oxide 2022; 125-126:1-11. [PMID: 35660109 DOI: 10.1016/j.niox.2022.05.005] [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: 02/02/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022]
Abstract
Impaired endogenous nitric oxide (NO) production may contribute to graft failure and premature mortality in kidney transplant recipients (KTR). We investigated potential associations of 24-h urinary NOx (NO3- + NO2-) excretion (uNOx) with long-term outcomes. uNOx was determined by HPLC and GC-MS in 698 KTR and in 132 kidney donors before and after donation. Additionally, we measured urinary nitroso species (RXNO) by gas-phase chemiluminescence. Median uNOx was lower in KTR compared to kidney donors (688 [393-1076] vs. 1301 [868-1863] before donation and 1312 [982-1853] μmol/24h after donation, P < 0.001). During median follow-up of 5.4 [4.8-6.1] years, 150 KTR died (61 due to cardiovascular disease) and 83 experienced graft failure. uNOx was inversely associated with all-cause mortality (HR per doubling of uNOx: 0.84 [95% CI 0.75-0.93], P < 0.001) and cardiovascular mortality (HR 0.78 [95% CI 0.67-0.92], P = 0.002). The association of uNOx with graft failure was lost when adjusted for renal function (HR per doubling of uNOx: 0.89 [95% CI 0.76-1.05], P = 0.17). There were no significant associations of urinary RXNO with outcomes. Our study suggests that KTR have lower NO production than healthy subjects and that lower uNOx is associated with a higher risk of all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Hanno Maassen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - M Yusof Said
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Anne-Roos S Frenay
- Department of Gynecology and Obstetrics, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Anne Koning
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Adrian Post
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kathrin Drabert
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Bernadette O Fernandez
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Reinold O B Gans
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Else van den Berg
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
26
|
Baudoux T, Jadot I, Declèves AE, Antoine MH, Colet JM, Botton O, De Prez E, Pozdzik A, Husson C, Caron N, Nortier JL. Experimental Aristolochic Acid Nephropathy: A Relevant Model to Study AKI-to-CKD Transition. Front Med (Lausanne) 2022; 9:822870. [PMID: 35602498 PMCID: PMC9115860 DOI: 10.3389/fmed.2022.822870] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/24/2022] [Indexed: 12/02/2022] Open
Abstract
Aristolochic acid nephropathy (AAN) is a progressive tubulointerstitial nephritis caused by the intake of aristolochic acids (AA) contained in Chinese herbal remedies or contaminated food. AAN is characterized by tubular atrophy and interstitial fibrosis, characterizing advanced kidney disease. It is established that sustained or recurrent acute kidney injury (AKI) episodes contribute to the progression of CKD. Therefore, the study of underlying mechanisms of AA-induced nephrotoxicity could be useful in understanding the complex AKI-to-CKD transition. We developed a translational approach of AKI-to-CKD transition by reproducing human AAN in rodent models. Indeed, in such models, an early phase of acute tubular necrosis was rapidly followed by a massive interstitial recruitment of activated monocytes/macrophages followed by cytotoxic T lymphocytes, resulting in a transient AKI episode. A later chronic phase was then observed with progressive tubular atrophy related to dedifferentiation and necrosis of tubular epithelial cells. The accumulation of vimentin and αSMA-positive cells expressing TGFβ in interstitial areas suggested an increase in resident fibroblasts and their activation into myofibroblasts resulting in collagen deposition and CKD. In addition, we identified 4 major actors in the AKI-to-CKD transition: (1) the tubular epithelial cells, (2) the endothelial cells of the interstitial capillary network, (3) the inflammatory infiltrate, and (4) the myofibroblasts. This review provides the most comprehensive and informative data we were able to collect and examines the pending questions.
Collapse
Affiliation(s)
- Thomas Baudoux
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Inès Jadot
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Anne-Emilie Declèves
- Laboratory of Molecular Biology, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Marie-Hélène Antoine
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jean-Marie Colet
- Department of Human Biology & Toxicology, Faculty of Medicine and Pharmacy, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Olivia Botton
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Eric De Prez
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Agnieszka Pozdzik
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Cécile Husson
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathalie Caron
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), Namur, Belgium
| | - Joëlle L Nortier
- Laboratory of Experimental Nephrology, Faculty of Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
27
|
Hudkova O, Krysiuk I, Drobot L, Latyshko N. Rhabdomyolysis attenuates activity of semicarbazide sensitive amine oxidase as the marker of nephropathy in diabetic rats. UKRAINIAN BIOCHEMICAL JOURNAL 2022. [DOI: 10.15407/ubj94.01.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
28
|
Zeng L, Liu Z, Zhou L, Chen M, Zheng X, Yang P, Zhao X, Tian Z. Effects of almonds on ameliorating salt-induced hypertension in dahl salt-sensitive rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:2710-2722. [PMID: 34708426 DOI: 10.1002/jsfa.11611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Excessive dietary salt intake is related to an increased risk of hypertension. Dietary functional foods probably could help to improve salt-induced hypertension. In this study, Dahl salt-sensitive (DSS) rats were used to investigate their metabolic differences from those of salt-resistant SS.13BN rats and determine whether dietary protein-rich almonds could ameliorate salt-induced elevation of blood pressure in DSS rats. RESULTS After high-salt intake, the systolic blood pressure and mean arterial pressure of the DSS rats increased dramatically. Metabolomics analysis indicated abnormal amino acid metabolism in their kidneys. Their renal nitric oxide (NO) content and nitric oxide synthase activity decreased significantly after high-salt diet. Oxidative stress also occurred in DSS rats. After the DSS rats received almond supplementation, the levels of various amino acids in their kidney increased, and renal arginine and NO contents were upregulated. Their renal hydrogen peroxide and malonaldehyde levels decreased, whereas renal catalase, superoxide dismutase and glutathione peroxidase activities and glutathione levels increased. CONCLUSION The renal abnormal amino acid metabolism of DSS rats contributed to the impaired NO production in response to high-salt intake. Together with salt-induced oxidative stress, high-salt diet intake ultimately led to an increase in the blood pressure of DSS rats. Protein-rich almond supplementation might prevent the development of salt-induced hypertension by restoring arginine and NO regeneration and alleviating salt-induced oxidative stress. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Li Zeng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zerong Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Luxin Zhou
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Meng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xuewei Zheng
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xinrui Zhao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Zhongmin Tian
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
29
|
Mavrakanas TA, Soomro QH, Charytan DM. Hydralazine-Isosorbide Dinitrate Use In Patients With End-Stage Kidney Disease On Dialysis. Kidney Int Rep 2022; 7:1332-1340. [PMID: 35685328 PMCID: PMC9171697 DOI: 10.1016/j.ekir.2022.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 10/25/2022] Open
|
30
|
Pereira BP, do Vale GT, Ceron CS. The role of nitric oxide in renovascular hypertension: from the pathophysiology to the treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:121-131. [PMID: 34994823 DOI: 10.1007/s00210-021-02186-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022]
Abstract
Renovascular hypertension is one of the most relevant causes of secondary hypertension, mostly caused by atherosclerotic renovascular stenosis or fibromuscular dysplasia. The increase in angiotensin II production, oxidative stress, and formation of peroxynitrite promotes the decrease in nitric oxide (NO) availability and the development of hypertension, renal and endothelial dysfunction, and cardiac and vascular remodeling. The NO produced by nitric oxide synthases (NOS) acts as a vasodilator; however, endothelial NOS uncoupling (eNOS) also contributes to NO reduced availability in renovascular hypertension. NO donors and NO-derived metabolites have been investigated in experimental renovascular hypertension and have shown promissory effects in attenuating blood pressure and organ damage in this condition. Therefore, understanding the role of decreased NO in the pathophysiology of renovascular hypertension promotes the study and development of NO donors and molecules that can be converted into NO (such as nitrate and nitrite), contributing for the treatment of this condition in the future.
Collapse
Affiliation(s)
- Bruna Pinheiro Pereira
- Departamento de Alimentos E Medicamentos, Universidade Federal de Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Gabriel Tavares do Vale
- Departamento de Ciências Biomédicas E da Saúde, Universidade Do Estado de Minas Gerais (UEMG), Belo Horizonte, Minas Gerais, Brazil
| | - Carla Speroni Ceron
- Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brasil.
| |
Collapse
|
31
|
Hu L, Chen Y, Zhou X, Hoek M, Cox J, Lin K, Liu Y, Blumenschein W, Grein J, Swaminath G. Effects of soluble guanylate cyclase stimulator on renal function in ZSF-1 model of diabetic nephropathy. PLoS One 2022; 17:e0261000. [PMID: 35085251 PMCID: PMC8794189 DOI: 10.1371/journal.pone.0261000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/23/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Diabetic nephropathy is associated with endothelial dysfunction and oxidative stress, in which the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling pathway is impaired. We hypothesize that sGC stimulator Compound 1 can enhance NO signaling, reduce proteinuria in a diabetic nephropathy preclinical model with diminished NO bioavailability and increased oxidized sGC. Therefore, we evaluated the effect of sGC stimulator Compound 1 on the renal effect in obese ZSF1 (ZSF1 OB) rats. MATERIALS AND METHODS The sGC stimulator Compound 1, the standard of care agent Enalapril, and a combination of Compound 1 and Enalapril were administered chronically to obese ZSF1 rats for 6 months. Mean arterial pressure, heart rate, creatinine clearance for glomerular filtration rate (eGFR), urinary protein excretion to creatinine ratio (UPCR), and urinary albumin excretion ratio (UACR) were determined during the study. The histopathology of glomerular and interstitial lesions was assessed at the completion of the study. RESULTS While both Compound 1 and Enalapril significantly reduced blood pressure, the combination of Compound 1 and Enalapril normalized blood pressure levels. Compound 1 improved eGFR and reduced UPCR and UACR. A combination of Enalapril and Compound 1 resulted in a marked reduction in UPCR and UACR and improved GFR. CONCLUSION The sGC stimulator Compound 1 as a monotherapy slowed renal disease progression, and a combination of the sGC stimulator with Enalapril provided greater renal protection in a rodent model of diabetic nephropathy.
Collapse
Affiliation(s)
- Lufei Hu
- Department of Cardiometabolic Diseases, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Yinhong Chen
- Department of Cardiometabolic Diseases, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Xiaoyan Zhou
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Maarten Hoek
- Department of Cardiometabolic Diseases, Merck & Co., Inc., Kenilworth, NJ, United States of America
- Biology Department, Maze Therapeutics, San Francisco, CA, United States of America
| | - Jason Cox
- Chemistry, Merck & Co., Inc., Kenilworth, NJ, United States of America
- Discovery Chemistry, Kinnate Biopharma, San Diego, CA, United States of America
| | - Ken Lin
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ, United States of America
- Drug Metabolism and Pharmacokinetics, BridgeBio, Palo Alto, CA, United States of America
| | - Yang Liu
- Department of Cardiometabolic Diseases, Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Wendy Blumenschein
- Department of Molecular Discovery Profiling and Expression, Merck & Co. Inc., Kenilworth, NJ, United States of America
| | - Jeff Grein
- Department of Molecular Discovery Profiling and Expression, Merck & Co. Inc., Kenilworth, NJ, United States of America
| | - Gayathri Swaminath
- Department of Cardiometabolic Diseases, Merck & Co., Inc., Kenilworth, NJ, United States of America
| |
Collapse
|
32
|
Forsse JS, Papadakis Z, Peterson MN, Taylor JK, Hess BW, Schwedock N, Allison DC, Griggs JO, Wilson RL, Grandjean PW. The Influence of an Acute Bout of Aerobic Exercise on Vascular Endothelial Function in Moderate Stages of Chronic Kidney Disease. Life (Basel) 2022; 12:life12010091. [PMID: 35054484 PMCID: PMC8781956 DOI: 10.3390/life12010091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/27/2021] [Accepted: 01/03/2022] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is directly influenced by the deleterious effects of systemic inflammation and oxidative stress. The vascular endothelium may transiently respond to aerobic exercise and improve post-exercise vascular renal function in moderate stages of CKD. Brachial artery flow-mediated dilation (FMD) is a nitric-oxide-dependent measure of endothelial function that is transiently potentiated by exercise. The purpose of the study was to determine the acute influence of a single bout of high-intensity interval exercise (HIIE) or steady-state moderate-intensity exercise (SSE) on endothelial dysfunction in moderate stages of CKD. Twenty participants (n = 6 men; n = 14 women) completed 30 min of SSE (65%) and HIIE (90:20%) of VO2reserve in a randomized crossover design. FMD measurements and blood samples were obtained before, 1 h, and 24 h post-exercise. FMD responses were augmented 1 h post-exercise in both conditions (p < 0.005). Relative to pre-exercise measures, total antioxidant capacity increased by 4.3% 24 h post-exercise (p = 0.012), while paraoxonase-1 was maintained 1 h and elevated by 6.1% 24 h after SSE, but not HIIE (p = 0.035). In summary, FMD can be augmented by a single episode of either HIIE or SSE in moderate stages of CKD. Modest improvements were observed in antioxidant analytes, and markers of oxidative stress were blunted in response to either SSE or HIIE.
Collapse
Affiliation(s)
- Jeffrey S. Forsse
- Department of Health Human Performance and Recreation, Baylor University, Waco, TX 76706, USA
- Correspondence: ; Tel.: +1-254-710-3900
| | - Zacharias Papadakis
- Department of Sport and Exercise Sciences, Barry University, Miami Shores, FL 33161, USA;
| | | | - James Kyle Taylor
- Clinical Laboratory Science, Auburn University-Montgomery, Montgomery, AL 36117, USA;
| | - Burritt W. Hess
- Waco Family Medicine, Waco, TX 76707, USA; (B.W.H.); (N.S.); (J.O.G.)
| | | | - Dale C. Allison
- Baylor Scott & White Health, Waco, TX 76712, USA; (D.C.A.); (R.L.W.)
| | - Jackson O. Griggs
- Waco Family Medicine, Waco, TX 76707, USA; (B.W.H.); (N.S.); (J.O.G.)
| | - Ronald L. Wilson
- Baylor Scott & White Health, Waco, TX 76712, USA; (D.C.A.); (R.L.W.)
| | - Peter W. Grandjean
- School of Applied Sciences, University of Mississippi, Oxford, MS 38677, USA;
| |
Collapse
|
33
|
Das S, Ray S, Devi T, Ghosh S, Harmalkar SS, Dhuri SN, Mondal P, Kumar P. Why Intermolecular Nitric Oxide (NO) Transfer? Exploring the Factors and Mechanistic Aspects of NO Transfer Reaction. Chem Sci 2022; 13:1706-1714. [PMID: 35282634 PMCID: PMC8827119 DOI: 10.1039/d1sc06803b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022] Open
Abstract
Small molecule activation & their transfer reactions in biological or catalytic reactions are greatly influenced by the metal-centers and the ligand frameworks. Here, we report the metal-directed nitric oxide (NO)...
Collapse
Affiliation(s)
- Sandip Das
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati 517507 India
| | - Soumyadip Ray
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati 517507 India
| | - Tarali Devi
- Humboldt-Universität zu Berlin, Institut für Chemie Brook-Taylor-Straße 2 D-12489 Berlin Germany
| | - Somnath Ghosh
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati 517507 India
| | | | - Sunder N Dhuri
- School of Chemical Sciences, Goa University Goa-403206 India
| | - Padmabati Mondal
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati 517507 India
| | - Pankaj Kumar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati 517507 India
| |
Collapse
|
34
|
Wu Q, Wang J, Wang Y, Xiang L, Tan Y, Feng J, Zhang Z, Zhang L. Targeted delivery of celastrol to glomerular endothelium and podocytes for chronic kidney disease treatment. NANO RESEARCH 2021; 15:3556-3568. [PMID: 34925707 PMCID: PMC8666268 DOI: 10.1007/s12274-021-3894-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 06/14/2023]
Abstract
UNLABELLED The etiology of chronic kidney disease (CKD) is complex and diverse, which could be briefly categorized to glomerular- or tubular-originated. However, the final outcomes of CKD are mainly glomerular sclerosis, endothelial dysfunction and injury, and chronic inflammation. Thus, targeted delivery of drugs to the glomeruli in order to ameliorate glomerular endothelial damage may help alleviate CKD and help enrich our knowledge. The herb tripterygium wilfordii shows therapeutic effect on kidney disease, and celastrol (CLT) is one of its active ingredients but with strong toxicity. Therefore, based on the unique structure and pathological characteristics of the glomerulus, we designed a targeted delivery system named peptides coupled CLT-phospholipid lipid nanoparticles (PC-PLNs) to efficiently deliver CLT to damaged endothelial cells and podocytes in the glomerulus for CKD treatment and research. PC-PLNs could effectively inhibit inflammation, reduce endothelial damage, alleviate CKD severity, and reduce the toxicity of CLT. We also studied the mechanism of CLT in the treatment of nephropathy and found that CLT can increase the level of NO by increasing eNOS while inhibiting the expression of VCAM-1, thus provides an anti-inflammatory effect. Therefore, our study not only offered an efficient CKD drug formulation for further development, but also provided new medical knowledge about CKD. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (attached with all the supporting tables and figures mentioned in this work) is available in the online version of this article at 10.1007/s12274-021-3894-x.
Collapse
Affiliation(s)
- Qingsi Wu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Jiading Wang
- College of Polymer Science and Engineering, Sichuan University, No. 24, South Block 1, First Ring Road, Chengdu, 610065 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Yuanfang Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Ling Xiang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Yulu Tan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Jiaxing Feng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, No. 24, South Block 1, First Ring Road, Chengdu, 610065 China
- Med-X Center for Materials, Sichuan University, No. 14 Section 3 South Renmin Road, Jinjiang District, Chengdu, 610000 China
| |
Collapse
|
35
|
Repurposing Riociguat to Target a Novel Paracrine Nitric Oxide-TRPC6 Pathway to Prevent Podocyte Injury. Int J Mol Sci 2021; 22:ijms222212485. [PMID: 34830371 PMCID: PMC8621407 DOI: 10.3390/ijms222212485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increased expression and activity of the Ca2+ channel transient receptor potential channel 6 (TRPC6) is associated with focal segmental glomerulosclerosis, but therapeutic strategies to target TRPC6 are currently lacking. Nitric oxide (NO) is crucial for normal glomerular function and plays a protective role in preventing glomerular diseases. We investigated if NO prevents podocyte injury by inhibiting injurious TRPC6-mediated signaling in a soluble guanylate cyclase (sGC)-dependent manner and studied the therapeutic potential of the sGC stimulator Riociguat. Experiments were performed using human glomerular endothelial cells and podocytes. Podocyte injury was induced by Adriamycin incubation for 24 h, with or without the NO-donor S-Nitroso-N-acetyl-DL-penicillamine (SNAP), the sGC stimulator Riociguat or the TRPC6 inhibitor Larixyl Acetate (LA). NO and Riociguat stimulated cGMP synthesis in podocytes, decreased Adriamycin-induced TRPC6 expression, inhibited the Adriamycin-induced TRPC6-mediated Ca2+ influx and reduced podocyte injury. The protective effects of Riociguat and NO were blocked when sGC activity was inhibited with 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or when TRPC6 activity was inhibited by LA. Our data demonstrate a glomerular (e)NOS-NO-sGC-cGMP-TRPC6 pathway that prevents podocyte injury, which can be translated to future clinical use by, e.g., repurposing the market-approved drug Riociguat.
Collapse
|
36
|
Ghadimi K, Cappiello J, Cooter-Wright M, Haney JC, Reynolds JM, Bottiger BA, Klapper JA, Levy JH, Hartwig MG. Inhaled Pulmonary Vasodilator Therapy in Adult Lung Transplant: A Randomized Clinical Trial. JAMA Surg 2021; 157:e215856. [PMID: 34787647 DOI: 10.1001/jamasurg.2021.5856] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Inhaled nitric oxide (iNO) is commonly administered for selectively inhaled pulmonary vasodilation and prevention of oxidative injury after lung transplant (LT). Inhaled epoprostenol (iEPO) has been introduced worldwide as a cost-saving alternative to iNO without high-grade evidence for this indication. Objective To investigate whether the use of iEPO will lead to similar rates of severe/grade 3 primary graft dysfunction (PGD-3) after adult LT when compared with use of iNO. Design, Setting, and Participants This health system-funded, randomized, blinded (to participants, clinicians, data managers, and the statistician), parallel-designed, equivalence clinical trial included 201 adult patients who underwent single or bilateral LT between May 30, 2017, and March 21, 2020. Patients were grouped into 5 strata according to key prognostic clinical features and randomized per stratum to receive either iNO or iEPO at the time of LT via 1:1 treatment allocation. Interventions Treatment with iNO or iEPO initiated in the operating room before lung allograft reperfusion and administered continously until cessation criteria met in the intensive care unit (ICU). Main Outcomes and Measures The primary outcome was PGD-3 development at 24, 48, or 72 hours after LT. The primary analysis was for equivalence using a two one-sided test (TOST) procedure (90% CI) with a margin of 19% for between-group PGD-3 risk difference. Secondary outcomes included duration of mechanical ventilation, hospital and ICU lengths of stay, incidence and severity of acute kidney injury, postoperative tracheostomy placement, and in-hospital, 30-day, and 90-day mortality rates. An intention-to-treat analysis was performed for the primary and secondary outcomes, supplemented by per-protocol analysis for the primary outcome. Results A total of 201 randomized patients met eligibility criteria at the time of LT (129 men [64.2%]). In the intention-to-treat population, 103 patients received iEPO and 98 received iNO. The primary outcome occurred in 46 of 103 patients (44.7%) in the iEPO group and 39 of 98 (39.8%) in the iNO group, leading to a risk difference of 4.9% (TOST 90% CI, -6.4% to 16.2%; P = .02 for equivalence). There were no significant between-group differences for secondary outcomes. Conclusions and Relevance Among patients undergoing LT, use of iEPO was associated with similar risks for PGD-3 development and other postoperative outcomes compared with the use of iNO. Trial Registration ClinicalTrials.gov identifier: NCT03081052.
Collapse
Affiliation(s)
- Kamrouz Ghadimi
- Department of Anesthesiology & Critical Care, Duke University School of Medicine, Durham, North Carolina
| | - Jhaymie Cappiello
- Department of Respiratory Care Services, Duke University Medical Center, Durham, North Carolina
| | - Mary Cooter-Wright
- Department of Anesthesiology & Critical Care, Duke University School of Medicine, Durham, North Carolina
| | - John C Haney
- Department of Surgery, Thoracic Transplant Surgery, Duke University School of Medicine, Durham, North Carolina
| | - John M Reynolds
- Department of Medicine, Transplant Pulmonology, Duke University School of Medicine, Durham, North Carolina
| | - Brandi A Bottiger
- Department of Anesthesiology & Critical Care, Duke University School of Medicine, Durham, North Carolina
| | - Jacob A Klapper
- Department of Surgery, Thoracic Transplant Surgery, Duke University School of Medicine, Durham, North Carolina
| | - Jerrold H Levy
- Department of Anesthesiology & Critical Care, Duke University School of Medicine, Durham, North Carolina
| | - Matthew G Hartwig
- Department of Surgery, Thoracic Transplant Surgery, Duke University School of Medicine, Durham, North Carolina
| | | |
Collapse
|
37
|
Hsiao PJ, Chiu CC, Tsai DJ, Ko PS, Chen YK, Cheng H, Su W, Lu KC, Su SL. Association between nitric oxide synthase T-786C genetic polymorphism and chronic kidney disease: Meta-analysis incorporating trial sequential analysis. PLoS One 2021; 16:e0258789. [PMID: 34662360 PMCID: PMC8523046 DOI: 10.1371/journal.pone.0258789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/05/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Several meta-analyses of the relationship between endothelial nitric oxide synthase (eNOS) T-786C gene polymorphism and chronic kidney disease (CKD) have been published. However, the results of these studies were inconsistent, and it is undetermined whether sample sizes are sufficient to reach a definite conclusion. OBJECTIVE To elucidate the relationship between T-786C and CKD by combining previous studies with our case-control sample and incorporate trial sequential analysis (TSA) to verify whether the sample size is adequate to draw a definite conclusion. METHODS PubMed and Embase databases were searched for relevant articles on eNOS T-786C and CKD before February 28, 2021. TSA was also incorporated to ascertain a conclusion. A total of 558 hemodialysis cases in the case-control study was recruited from nine dialysis centers in the northern area of Taiwan in 2020. Additionally, 640 healthy subjects of the control group, with estimated glomerular filtration rate (eGFR) ≥ 60 mL/min/1.73 m2, were selected from participants of the annual elderly health examination program at the Tri-Service General Hospital. The functional analysis was based on eQTL data from GTExPortal. RESULTS After screening with eligibility criteria, 15 papers were included and eventually combined in a meta-analysis. The result of the TSA showed that the sample size for Caucasians was adequate to ascertain the correlation between eNOS T-786C and CKD but was insufficient for Asians. Therefore, we added our case-control samples (n = 1198), though not associated with CKD (odds ratio [OR] = 1.01, 95% confidence interval [CI] = 0.69-1.46), into a meta-analysis, which supported that eNOS T-786C was significantly associated with CKD in Asians (OR = 1.39, 95% CI = 1.04-1.85) by using an adequate cumulative sample size (n = 4572) analyzed by TSA. Data of eQTL from GTEx showed that T-786C with the C minor allele exhibited relatively lower eNOS mRNA expression in whole blood, indicating the hazardous role of eNOS T-786C in CKD. CONCLUSIONS eNOS T-786C genetic polymorphism was of conclusive significance in the association with CKD among Asians in our meta-analysis. Our case-control samples play a decisive role in changing conclusions from indefinite to definite.
Collapse
Affiliation(s)
- Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan, R.O.C.
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.
- Department of Life Sciences, National Central University, Taoyuan, Taiwan, R.O.C.
- Big Data Research Center, Fu-Jen Catholic University, New Taipei City, Taiwan, R.O.C.
| | - Chih-Chien Chiu
- Division of Infectious Diseases, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, National Defense Medical Center, Taoyuan, Taiwan, R.O.C.
| | - Dung-Jang Tsai
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Pi-Shao Ko
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Ying-Kai Chen
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C.
- Division of Nephrology, Department of Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan, R.O.C.
| | - Hao Cheng
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Wen Su
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, R.O.C.
| | - Sui-Lung Su
- School of Public Health, National Defense Medical Center, Taipei, Taiwan, R.O.C.
| |
Collapse
|
38
|
Renoprotection Induced by Aerobic Training Is Dependent on Nitric Oxide Bioavailability in Obese Zucker Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3683796. [PMID: 34621463 PMCID: PMC8492245 DOI: 10.1155/2021/3683796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/21/2021] [Indexed: 11/21/2022]
Abstract
Aerobic training (AT) promotes several health benefits that may attenuate the progression of obesity associated diabetes. Since AT is an important nitric oxide (NO−) inducer mediating kidney-healthy phenotype, the present study is aimed at investigating the effects of AT on metabolic parameters, morphological, redox balance, inflammatory profile, and vasoactive peptides in the kidney of obese-diabetic Zucker rats receiving L-NAME (N(omega)-nitro-L-arginine methyl ester). Forty male Zucker rats (6 wk old) were assigned into four groups (n = 10, each): sedentary lean rats (CTL-Lean), sedentary obese rats (CTL-Obese), AT trained obese rats without blocking nitric oxide synthase (NOS) (Obese+AT), and obese-trained with NOS block (Obese+AT+L-NAME). AT groups ran 60 min in the maximal lactate steady state (MLSS), five days/wk/8 wk. Obese+AT rats improved glycemic homeostasis, SBP, aerobic capacity, renal mitochondria integrity, redox balance, inflammatory profile (e.g., TNF-α, CRP, IL-10, IL-4, and IL-17a), and molecules related to renal NO− metabolism (klotho/FGF23 axis, vasoactive peptides, renal histology, and reduced proteinuria). However, none of these positive outcomes were observed in CTL-Obese and Obese+AT+L-NAME (p < 0.0001) groups. Although Obese+AT+L-NAME lowered BP (compared with CTL-Obese; p < 0.0001), renal damage was observed after AT intervention. Furthermore, AT training under conditions of low NO− concentration increased signaling pathways associated with ACE-2/ANG1-7/MASr. We conclude that AT represents an important nonpharmacological intervention to improve kidney function in obese Zucker rats. However, these renal and metabolic benefits promoted by AT are dependent on NO− bioavailability and its underlying regulatory mechanisms.
Collapse
|
39
|
Farahmand M, Ramezani Tehrani F, Khalili D, Cheraghi L, Azizi F. Endogenous estrogen exposure and chronic kidney disease; a 15-year prospective cohort study. BMC Endocr Disord 2021; 21:155. [PMID: 34348694 PMCID: PMC8336110 DOI: 10.1186/s12902-021-00817-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 07/18/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Despite strong evidence demonstrating the role of estrogen as a protective factor for kidney function in women, limited data are available regarding the influence of endogenous estrogen exposure (EEE) on chronic kidney disease (CKD). The present study aimed to assess the incidence of CKD in women with various levels of EEE. METHODS In a prospective population-based study over a 15-year follow-up, a total of 3043 eligible women aged 30-70 years, participating in Tehran-Lipid and Glucose-Study were recruited and divided into two groups (EEE < 11 and EEE ≥ 11 years). EEE calculated based on age at menarche, age at menopause, number and duration of pregnancies, lactation, and duration of oral contraceptive use after excluding the progesterone dominant phase of the menstrual cycle. Cox's proportional hazards model was applied to estimate the hazard ratio of CKD between the study groups, after adjusting for confounders. RESULTS The total cumulative incidence rate of CKD was 50.1 per 1000 person years; 95% CI: 47.7-52.6); this was 53.9 (95%CI, 50.2-57.8) and 47.1 (95%CI, 44.0-50.4) per 1000 person years in women with EEE < 11 and EEE ≥ 11 years, respectively. The model adjusted for age, BMI, smoking, hypertension, and diabetes showed that the hazard ratio (HR) of incidence CKD in women with EEE < 11 compare to those with EEE ≥ 11 years in the subgroup of women aged< 45 years was 2.66(95% CI, 2.2, 3.2), whereas, in the subgroup aged ≥45 years, it was 1.22 (95% CI, 1.04, 1.4). CONCLUSION This study shows a higher HR of CKD incidence in women with low EEE levels in their later life. Screening of these women for CKD may be recommended.
Collapse
Affiliation(s)
- Maryam Farahmand
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Ramezani Tehrani
- Reproductive Endocrinology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Davood Khalili
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Epidemiology and Biostatistics, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leila Cheraghi
- Department of Epidemiology and Biostatistics, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Wang Q, Liu L, Jonas JB, Gao B, Wu SL, Chen SH, Yan YN, Yang JY, Zhou WJ, Yang MC, Chan SY, Zhang L, Wei WB, Wang YX. Albuminuria and retinal vessel density in diabetes without diabetic retinopathy: the Kailuan Eye Study. Acta Ophthalmol 2021; 99:e669-e678. [PMID: 33354932 DOI: 10.1111/aos.14670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Diabetic retinopathy and chronic kidney disease are both major complications of diabetes mellitus. We explored the relationship between retinal vessel density (VD) and albuminuria in diabetic patients without conventionally defined diabetic retinopathy. METHODS The cross-sectional community-based Kailuan Diabetic Retinopathy Study included patients with type 2 diabetes without diabetic retinopathy who participated in the community-based longitudinal Kailuan study and who had undergone ocular fundus photography, kidney function assessment, and optical coherence tomographic angiography (OCT-angiography) for the assessment the retinal perfusion density (PD) and retinal VD. RESULTS The study included 447 patients (mean age: 60.9 ± 9.7 years). Higher PD and VD were associated with a lower urinary albumin-creatinine ratio (uACR) (macular region: p = 0.007: standardized regression coefficient beta: -0.14; and p = 0.008, beta: -0.13, respectively; parafoveal region: p = 0.006, beta: -0.14; and p = 0.007, beta: -0.14, respectively) after adjusting for age and ocular axial length. In a reverse manner, higher uACR was associated with lower PD and VD (macular region: p = 0.009, beta: -0.14; and p = 0.01, beta: -0.14, respectively; parafoveal region: p = 0.008, beta: -0.14; and p = 0.01, beta: -0.14, respectively), after adjusting for diabetes duration, blood pressure, serum concentration of C-reactive protein and high-density lipoprotein cholesterol and ocular axial length. In a multivariable model, the prevalence of macroalbuminuria increased by 11% (95% CI: 2%, 18%) and 17% (95% CI: 3%, 30%), respectively, for each mm-1 decrease in VD and each unit decrease in PD. CONCLUSIONS AND RELEVANCE After adjusting for systemic and ocular parameters, diabetic patients without diabetic retinopathy showed a reduction in OCT-angiographic retinal vascular measurements in association with systemic parameters indicating chronic kidney disease. Optical coherence tomographic (OCT)-angiographic retinal microvascular parameters may serve as markers for chronic kidney disease.
Collapse
Affiliation(s)
- Qian Wang
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| | - Lili Liu
- Renal Division Department of Medicine Peking University First Hospital Beijing China
| | - Jost B. Jonas
- Department of Ophthalmology Medical Faculty Mannheim of the Ruprecht‐Karis‐University Mannheim Germany
| | - Bixia Gao
- Renal Division Department of Medicine Peking University First Hospital Beijing China
| | - Shou Ling Wu
- Cardiology Department Kailuan General Hospital Tangshan China
| | | | - Yan Ni Yan
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| | - Jing Yan Yang
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| | - Wen Jia Zhou
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| | - Mo Chi Yang
- Department of Ophthalmology General Hospital of Ningxia Medical University Ningxia China
| | - Szy Yann Chan
- Department of Ophthalmology Peking University Third Hospital Beijing China
| | - Luxia Zhang
- Renal Division Department of Medicine Peking University First Hospital Beijing China
- National Institute of Health Data Science at Peking University Beijing China
| | - Wen Bin Wei
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| | - Ya Xing Wang
- Beijing Ophthalmology and Visual Science Key Lab Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment Beijing Tongren Eye CenterBeijing Tongren HospitalCapital Medical University Beijing China
| |
Collapse
|
41
|
Verma S, Singh P, Khurana S, Ganguly NK, Kukreti R, Saso L, Rana DS, Taneja V, Bhargava V. Implications of oxidative stress in chronic kidney disease: a review on current concepts and therapies. Kidney Res Clin Pract 2021; 40:183-193. [PMID: 34078023 PMCID: PMC8237115 DOI: 10.23876/j.krcp.20.163] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/05/2021] [Indexed: 01/04/2023] Open
Abstract
Moderate levels of endogenous reactive oxygen species (ROS) are important for various cellular activities, but high levels lead to toxicity and are associated with various diseases. Levels of ROS are maintained as a balance between oxidants and antioxidants. Accumulating data suggest that oxidative stress is a major factor in deterioration of renal function. In this review, we highlight the possible mechanism by which oxidative stress can lead to chronic kidney disease (CKD). This review also describes therapies that counter the effect of oxidative stress in CKD patients. Numerous factors such as upregulation of genes involved in oxidative phosphorylation and ROS generation, chronic inflammation, vitamin D deficiency, and a compromised antioxidant defense mechanism system cause progressive detrimental effects on renal function that eventually lead to loss of kidney function. Patients with renal dysfunction are highly susceptible to oxidative stress, as risk factors such as diabetes, renal hypertension, dietary restrictions, hemodialysis, and old age predispose them to increased levels of ROS. Biomolecular adducts (DNA, proteins, and lipids) formed due to reaction with ROS can be used to determine oxidative stress levels. Based on the strong correlation between oxidative stress and CKD, reversal of oxidative stress is being explored as a major therapeutic option. Xanthine oxidase inhibitors, dietary antioxidants, and other agents that scavenge free radicals are gaining interest as treatment modalities in CKD patients.
Collapse
Affiliation(s)
- Sagar Verma
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Priyanka Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shiffali Khurana
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | | | - Ritushree Kukreti
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | | | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Vinant Bhargava
- Department of Nephrology, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
42
|
Endothelial-specific overexpression of cationic amino acid transporter-1 prevents loss of kidney function in heart failure. Clin Sci (Lond) 2021; 134:2755-2769. [PMID: 33034619 DOI: 10.1042/cs20200087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/17/2020] [Accepted: 10/08/2020] [Indexed: 12/21/2022]
Abstract
Heart failure (HF) is associated with impaired L-arginine transport. In the present study, we tested the hypothesis that augmented L-arginine transport prevents the loss of kidney function in HF. Renal function was assessed in wildtype mice (WT), transgenic mice with HF (dilated cardiomyopathy, DCM) and double transgenic mice (double transgenic mice with DCM and CAT-1 overexpression, HFCAT-1) with HF and endothelial-specific overexpression of the predominant L-arginine transporter, cationic amino acid transporter-1 (CAT-1) (n=4-8/group). Cardiac function was assessed via echocardiography and left ventricular catheterisation. Renal function was assessed via quantification of albuminuria and creatinine clearance. Plasma nitrate and nitrite levels together with renal fibrosis and inflammatory markers were also quantified at study end. Albumin/creatinine ratio was two-fold greater in DCM mice than in WT mice (P=0.002), and tubulointerstitial and glomerular fibrosis were approximately eight- and three-fold greater, respectively, in DCM mice than in WT mice (P≤0.02). Critically, urinary albumin/creatinine ratio and tubulointerstitial and glomerular fibrosis were less in HFCAT-1 mice than in DCM mice (P<0.05). Renal CAT-1 expression and plasma nitrate and nitrite levels were less in DCM mice compared with WT (P≤0.03) but was greater in HFCAT-1 mice than in DCM mice (P≤0.009). Renal expression of IL-10 was less in DCM mice compared with WT (P<0.001) but was greater in HFCAT-1 mice compared with DCM mice (P=0.02). Our data provide direct evidence that augmented L-arginine transport prevents renal fibrosis, inflammation and loss of kidney function in HF.
Collapse
|
43
|
Arginine Metabolites as Biomarkers of Myocardial Ischaemia, Assessed with Cardiac Magnetic Resonance Imaging in Chronic Kidney Disease. Biomolecules 2021; 11:biom11030416. [PMID: 33799818 PMCID: PMC8002086 DOI: 10.3390/biom11030416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/24/2021] [Accepted: 03/06/2021] [Indexed: 12/28/2022] Open
Abstract
(1) Background: Cardiovascular disease (CVD) is the major cause of morbidity and mortality in patients with chronic kidney disease (CKD). Myocardial oxygenation and perfusion response to stress, using oxygen-sensitive cardiovascular magnetic resonance (OS-CMR) and stress T1 mapping respectively, are impaired in CKD patients with and without known coronary artery disease (CAD). Endothelial dysfunction, assessed by circulating levels of asymmetric dimethylarginine (ADMA) and homoarginine (HMA), promotes atherosclerosis. We hypothesized that in CKD patients, worsening endothelial dysfunction is associated with worsening myocardial oxygenation and perfusion as assessed by change in OS-CMR signal intensity (Δ OS-CMR SI) and stress T1 (ΔT1) values. (2) Methods: 38 patients with advanced CKD underwent cardiovascular magnetic resonance (CMR) scanning at 3 Tesla. OS-CMR and T1 mapping images were acquired both at rest and after adenosine stress and analyzed semi-quantitatively. Serum ADMA and HMA concentrations were assessed using mass spectrometry. (3) Results: There was no significant correlation between Δ OS-CMR SI and ADMA or HMA. Interestingly, there was a significant negative correlation seen between Δ T1 and ADMA (r = -0.419, p = 0.037, n = 30) but not between Δ T1 and HMA. (4) Conclusions: Stress T1 response is impaired in CKD patients and is independently associated with higher circulating ADMA concentrations.
Collapse
|
44
|
The Potential Role of Creatine in Vascular Health. Nutrients 2021; 13:nu13030857. [PMID: 33807747 PMCID: PMC7999364 DOI: 10.3390/nu13030857] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 12/11/2022] Open
Abstract
Creatine is an organic compound, consumed exogenously in the diet and synthesized endogenously via an intricate inter-organ process. Functioning in conjunction with creatine kinase, creatine has long been known for its pivotal role in cellular energy provision and energy shuttling. In addition to the abundance of evidence supporting the ergogenic benefits of creatine supplementation, recent evidence suggests a far broader application for creatine within various myopathies, neurodegenerative diseases, and other pathologies. Furthermore, creatine has been found to exhibit non-energy related properties, contributing as a possible direct and in-direct antioxidant and eliciting anti-inflammatory effects. In spite of the new clinical success of supplemental creatine, there is little scientific insight into the potential effects of creatine on cardiovascular disease (CVD), the leading cause of mortality. Taking into consideration the non-energy related actions of creatine, highlighted in this review, it can be speculated that creatine supplementation may serve as an adjuvant therapy for the management of vascular health in at-risk populations. This review, therefore, not only aims to summarize the current literature surrounding creatine and vascular health, but to also shed light onto the potential mechanisms in which creatine may be able to serve as a beneficial supplement capable of imparting vascular-protective properties and promoting vascular health.
Collapse
|
45
|
Soomro QH, Charytan DM. Cardiovascular autonomic nervous system dysfunction in chronic kidney disease and end-stage kidney disease: disruption of the complementary forces. Curr Opin Nephrol Hypertens 2021; 30:198-207. [PMID: 33395034 DOI: 10.1097/mnh.0000000000000686] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Several nontraditional risk factors have been the focus of research in an attempt to understand the disproportionately high cardiovascular morbidity and mortality in chronic kidney disease (CKD) and end-stage kidney disease (ESKD) populations. One such category of risk factors is cardiovascular autonomic dysfunction. Its true prevalence in the CKD/ESKD population is unknown but existing evidence suggests it is common. Due to lack of standardized diagnostic and treatment options, this condition remains undiagnosed and untreated in many patients. In this review, we discuss current evidence pointing toward the role of autonomic nervous system (ANS) dysfunction in CKD, building off of crucial historical evidence and thereby highlighting the areas in need for future research interest. RECENT FINDINGS There are several key mediators and pathways leading to cardiovascular autonomic dysfunction in CKD and ESKD. We review studies exploring the mechanisms involved and discuss the current measurement tools and indices to evaluate the ANS and their pitfalls. There is a strong line of evidence establishing the temporal sequence of worsening autonomic function and kidney function and vice versa. Evidence linking ANS dysfunction and arrhythmia, sudden cardiac death, intradialytic hypotension, heart failure and hypertension are discussed. SUMMARY There is a need for early recognition and referral of CKD and ESKD patients suspected of cardiovascular ANS dysfunction to prevent the downstream effects described in this review.There are many unknowns in this area and a clear need for further research.
Collapse
Affiliation(s)
- Qandeel H Soomro
- Nephrology Division, Department of Medicine, NYU Langone Medical Center, New York, New York, USA
| | | |
Collapse
|
46
|
Vila Cuenca M, Hordijk PL, Vervloet MG. Most exposed: the endothelium in chronic kidney disease. Nephrol Dial Transplant 2021; 35:1478-1487. [PMID: 31071222 PMCID: PMC7473805 DOI: 10.1093/ndt/gfz055] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence indicates that the pathological changes of the endothelium may contribute to the development of cardiovascular complications in chronic kidney disease (CKD). Non-traditional risk factors related to CKD are associated with the incidence of cardiovascular disease, but their role in uraemic endothelial dysfunction has often been disregarded. In this context, soluble α-Klotho and vitamin D are of importance to maintain endothelial integrity, but their concentrations decline in CKD, thereby contributing to the dysfunction of the endothelial lining. These hormonal disturbances are accompanied by an increment of circulating fibroblast growth factor-23 and phosphate, both exacerbating endothelial toxicities. Furthermore, impaired renal function leads to an increment of inflammatory mediators, reactive oxygen species and uraemic toxins that further aggravate the endothelial abnormalities and in turn also inhibit the regeneration of disrupted endothelial lining. Here, we highlight the distinct endothelial alterations mediated by the abovementioned non-traditional risk factors as demonstrated in experimental studies and connect these to pathological changes in CKD patients, which are driven by endothelial disturbances, other than atherosclerosis. In addition, we describe therapeutic strategies that may promote restoration of endothelial abnormalities by modulating imbalanced mineral homoeostasis and attenuate the impact of uraemic retention molecules, inflammatory mediators and reactive oxygen species. A clinical perspective on endothelial dysfunction in CKD may translate into reduced structural and functional abnormalities of the vessel wall in CKD, and ultimately improved cardiovascular disease.
Collapse
Affiliation(s)
- Marc Vila Cuenca
- Department of Nephrology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter L Hordijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc G Vervloet
- Department of Nephrology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Yamaguchi Y, Zampino M, Moaddel R, Chen TK, Tian Q, Ferrucci L, Semba RD. Plasma metabolites associated with chronic kidney disease and renal function in adults from the Baltimore Longitudinal Study of Aging. Metabolomics 2021; 17:9. [PMID: 33428023 PMCID: PMC9220986 DOI: 10.1007/s11306-020-01762-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Chronic kidney disease (CKD) is an important cause of disability and death, but its pathogenesis is poorly understood. Plasma metabolites can provide insights into underlying processes associated with CKD. OBJECTIVES To clarify the relationship of plasma metabolites with CKD and renal function in human. METHODS We used a targeted metabolomics approach to characterize the relationship of 450 plasma metabolites with CKD and estimated glomerular filtration rate (eGFR) in 616 adults, aged 38-94 years, who participated in the Baltimore Longitudinal Study of Aging. RESULTS There were 74 (12.0%) adults with CKD. Carnitine, acetylcarnitine, propionylcarnitine, butyrylcarnitine, trigonelline, trimethylamine N-oxide (TMAO), 1-methylhistidine, citrulline, homoarginine, homocysteine, sarcosine, symmetric dimethylarginine, aspartate, phenylalanine, taurodeoxycholic acid, 3-indolepropionic acid, phosphatidylcholines (PC).aa.C40:2, PC.aa.C40:3, PC.ae.C40:6, triglycerides (TG) 20:4/36:3, TG 20:4/36:4, and choline were associated with higher odds of CKD in multivariable analyses adjusting for potential confounders and using a false discovery rate (FDR) to address multiple testing. Six acylcarnitines, trigonelline, TMAO, 18 amino acids and biogenic amines, taurodeoxycholic acid, hexoses, cholesteryl esters 22:6, dehydroepiandrosterone sulfate, 3-indolepropionic acid, 2 PCs, 17 TGs, and choline were negatively associated with eGFR, and hippuric acid was positively associated with eGFR in multivariable analyses adjusting for potential confounders and using a FDR approach. CONCLUSION The metabolites associated with CKD and reduced eGFR suggest that several pathways, such as the urea cycle, the arginine-nitric oxide pathway, the polyamine pathway, and short chain acylcarnitine metabolism are altered in adults with CKD and impaired renal function.
Collapse
Affiliation(s)
- Yuko Yamaguchi
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Smith Building, M015, 400 N. Broadway, Baltimore, MD, 21287, USA.
| | - Marta Zampino
- National Institutes On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ruin Moaddel
- National Institutes On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Teresa K Chen
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Qu Tian
- National Institutes On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- National Institutes On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Smith Building, M015, 400 N. Broadway, Baltimore, MD, 21287, USA
| |
Collapse
|
48
|
Abstract
The prevalence of cardiovascular and metabolic disease coupled with kidney dysfunction is increasing worldwide. This triad of disorders is associated with considerable morbidity and mortality as well as a substantial economic burden. Further understanding of the underlying pathophysiological mechanisms is important to develop novel preventive or therapeutic approaches. Among the proposed mechanisms, compromised nitric oxide (NO) bioactivity associated with oxidative stress is considered to be important. NO is a short-lived diatomic signalling molecule that exerts numerous effects on the kidneys, heart and vasculature as well as on peripheral metabolically active organs. The enzymatic L-arginine-dependent NO synthase (NOS) pathway is classically viewed as the main source of endogenous NO formation. However, the function of the NOS system is often compromised in various pathologies including kidney, cardiovascular and metabolic diseases. An alternative pathway, the nitrate-nitrite-NO pathway, enables endogenous or dietary-derived inorganic nitrate and nitrite to be recycled via serial reduction to form bioactive nitrogen species, including NO, independent of the NOS system. Signalling via these nitrogen species is linked with cGMP-dependent and independent mechanisms. Novel approaches to restoring NO homeostasis during NOS deficiency and oxidative stress have potential therapeutic applications in kidney, cardiovascular and metabolic disorders.
Collapse
|
49
|
Charytan DM, Hsu JY, Mc Causland FR, Waikar SS, Ikizler TA, Raj DS, Landis JR, Mehrotra R, Williams M, DiCarli M, Skali H, Kimmel PL, Kliger AS, Dember LM. Combination Hydralazine and Isosorbide Dinitrate in Dialysis-Dependent ESRD (HIDE): A Randomized, Placebo-Controlled, Pilot Trial. KIDNEY360 2020; 1:1380-1389. [PMID: 35372900 DOI: 10.34067/kid.0004342020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/05/2020] [Indexed: 11/27/2022]
Abstract
Background Combination therapy with isosorbide dinitrate (ISD) and hydralazine (HY) reduces heart failure mortality. The safety and tolerability in individuals requiring maintenance hemodialysis (HD) is unknown. Methods Single-center, randomized, placebo-controlled, double-blind pilot trial to explore safety and tolerability of ISD/HY in maintenance HD. Participants were randomized to placebo or combination ISD/HY. Dose was escalated over 3 weeks from ISD 10 mg/HY 10 mg to ISD 40 mg/HY 75 mg three times per day with the maximum tolerated dose maintained for the subsequent 21 weeks. Primary endpoints included adverse events, adverse events precluding further treatment with study medication, serious hypotension (i.e., requiring hospitalization or emergency room visit), and recurrent intra-dialytic hypotension. Efficacy signals included change in mitral annular E' velocity by tissue Doppler echocardiography and change in left ventricular coronary flow reserve on positron emission tomography. Results A total of 17 individuals were randomized to ISD/HY (N=7) or placebo (N=10). All participants assigned to ISD/HY completed dose escalation to 40/75 mg, but dose reductions were required in two participants. No participants discontinued therapy. There were no serious hypotension events. Recurrent intradialytic hypotension was less frequent with ISD/HY (0.47 events/patient-year) than placebo (1.83 events/patient-year, P=0.04). In contrast, nausea (ISD/HY, 1.90 events/patient-year; placebo, 0.50 events/patient-year, P=0.03) was significantly more frequent, and headache and diarrhea were numerically but not significantly more frequent with ISD/HY. Adverse events were more frequent with ISD/HY (11.4 events/patient-year) than placebo (6.31 events/patient-year). We did not detect between-group differences in the change in E' (P=0.34); ISD/HY showed a mean increase of 0.6 cm/s (SD 1.1), and placebo showed a mean decrease of 0.04 cm/s (SD 0.9). Changes in coronary flow reserve were minimal, -0.3 (0.2) with ISD/HY and -0.03 (0.5) in the placebo group, P=0.19. Conclusions ISD/HY appears to be well tolerated in patients being treated with maintenance HD, but headache and gastrointestinal side effects occur more frequently with ISD/HY compared with placebo.
Collapse
Affiliation(s)
- David M Charytan
- Division of Nephrology, Department of Medicine, New York University Grossman School of Medicine, New York, New York
| | - Jesse Y Hsu
- Department of Biostatistics, Epidemiology and Informatics, and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Finnian R Mc Causland
- Renal Division, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Sushrut S Waikar
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts
| | - T Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, and Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dominic S Raj
- Division of Renal Diseases and Hypertension, George Washington University School of Medicine, Washington, DC
| | - J Richard Landis
- Department of Biostatistics, Epidemiology and Informatics, and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rajnish Mehrotra
- Division of Nephrology, Department of Medicine, Kidney Research Institute, University of Washington, Seattle, Washington
| | - Mark Williams
- Renal Division, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Marcelo DiCarli
- Departments of Radiology and Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Hicham Skali
- Cardiovascular Division, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Paul L Kimmel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Alan S Kliger
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Laura M Dember
- Renal, Electrolyte and Hypertension Division, and Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | |
Collapse
|
50
|
Kelly DM, Rothwell PM. Impact of multimorbidity on risk and outcome of stroke: Lessons from chronic kidney disease. Int J Stroke 2020; 16:758-770. [PMID: 33243088 PMCID: PMC8521355 DOI: 10.1177/1747493020975250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
With both an aging population and greater post-stroke survival, multimorbidity is a growing healthcare challenge, affecting over 40% of stroke patients, and rising rapidly and predictably with increasing age. Commonly defined as the co-occurrence of two or more chronic conditions, multimorbidity burden is a strong adverse prognostic factor, associated with greater short- and long-term stroke mortality, worse rehabilitation outcomes, and reduced use of secondary prevention. Chronic kidney disease can be considered as the archetypal comorbidity, being age-dependent and also affecting about 40% of stroke patients. Chronic kidney disease and stroke share very similar traditional cardiovascular risk factor profiles such as hypertension and diabetes, though novel chronic kidney disease-specific risk factors such as inflammation and oxidative stress have also been proposed. Using chronic kidney disease as an exemplar condition, we explore the mechanisms of risk in multimorbidity, implications for management, impact on stroke severity, and downstream consequences such as post-stroke cognitive impairment and dementia.
Collapse
Affiliation(s)
- Dearbhla M Kelly
- Nuffield Department of Clinical Neurosciences, Wolfson Centre for Prevention of Stroke and Dementia, John Radcliffe Hospital, University of Oxford, UK
| | - Peter M Rothwell
- Nuffield Department of Clinical Neurosciences, Wolfson Centre for Prevention of Stroke and Dementia, John Radcliffe Hospital, University of Oxford, UK
| |
Collapse
|