1
|
Zhong F, Li W, Zhao C, Jin L, Lu X, Zhao Y, Pu J, Ge H. Basigin Deficiency Induces Spontaneous Polycystic Kidney in Mice. Hypertension 2024; 81:114-125. [PMID: 37955149 DOI: 10.1161/hypertensionaha.123.21486] [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: 05/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Polycystic kidney disease is the most common hereditary kidney disorder with early and frequent hypertension symptoms. The mechanisms of cyst progression in polycystic kidney disease remain incompletely understood. METHODS Bsg (basigin) heterozygous and homozygous knockout mice were generated using cas9 system, and Bsg overexpression was achieved by adeno-associated virus serotype 9 injection. Renal morphology was investigated through histological and imaging analysis. Molecular analysis was performed through transcriptomic profiling and biochemical approaches. RESULTS Bsg-deficient mice exhibited significantly elevated arterial blood pressure. Further investigation demonstrated that Bsg deficiency triggers spontaneous cystic formation in mouse kidneys, which shares similar cyst pathological features and common transcriptional regulatory pathways with human polycystic kidney disease. Moreover, Bsg disruption promoted polycystin-1 ubiquitination and degradation, leading to activation of polycystic kidney disease associated cAMP and AMPK signaling pathways in Bsg knockout mouse kidneys. Finally, adeno-associated virus serotype 9 mediated Bsg reexpression reversed cystic progression in Bsg knockout mice in vivo, and Bsg overexpression inhibited the expansion of Madin-Darby canine kidney cysts in vitro. CONCLUSIONS Our findings show that Bsg deficiency leads to an early-onset spontaneous polycystic kidney phenotype, suggesting that dysregulated Bsg signaling may be a contributing factor in cystogenesis.
Collapse
Affiliation(s)
- Fangyuan Zhong
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Wenli Li
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Chenxu Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Lixing Jin
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Xiyuan Lu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| | - Heng Ge
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
2
|
Zhu J, Liu F, Mao J. Clinical findings, underlying pathogenetic processes and treatment of vascular dysfunction in autosomal dominant polycystic kidney disease. Ren Fail 2023; 45:2282027. [PMID: 37970664 PMCID: PMC11001366 DOI: 10.1080/0886022x.2023.2282027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited disorder characterized by the development of fluid-filled cysts in the kidneys. The primary cause of ADPKD is mutations in the PKD1 (polycystic kidney disease 1) or PKD2 (polycystic kidney disease 2) gene. Patients with ADPKD often develop a variety of vascular abnormalities, which have a major impact on the structure and function of the blood vessels and can lead to complications such as hypertension, intracranial aneurysm (ICAN), and atherosclerosis. The progression of ADPKD involves intricate molecular and cellular processes that lead to the development of these vascular abnormalities. Our understanding of these processes remains incomplete, and available treatment options are limited. The aim of this review is to delve into the underlying mechanisms of these vascular abnormalities and to explore potential interventions.
Collapse
Affiliation(s)
- Jinjun Zhu
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
3
|
Sagar PS, Rangan GK. Cardiovascular Manifestations and Management in ADPKD. Kidney Int Rep 2023; 8:1924-1940. [PMID: 37850017 PMCID: PMC10577330 DOI: 10.1016/j.ekir.2023.07.017] [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: 03/16/2023] [Revised: 06/27/2023] [Accepted: 07/24/2023] [Indexed: 10/19/2023] Open
Abstract
Cardiovascular disease (CVD) is the major cause of mortality in autosomal dominant polycystic kidney disease (ADPKD) and contributes to significant burden of disease. The manifestations are varied, including left ventricular hypertrophy (LVH), intracranial aneurysms (ICAs), valvular heart disease, and cardiomyopathies; however, the most common presentation and a major modifiable risk factor is hypertension. The aim of this review is to detail the complex pathogenesis of hypertension and other extrarenal cardiac and vascular conditions in ADPKD drawing on preclinical, clinical, and epidemiological evidence. The main drivers of disease are the renin-angiotensin-aldosterone system (RAAS) and polycystin-related endothelial cell dysfunction, with the sympathetic nervous system (SNS), nitric oxide (NO), endothelin-1 (ET-1), and asymmetric dimethylarginine (ADMA) likely playing key roles in different disease stages. The reported rates of some manifestations, such as LVH, have decreased likely due to the use of antihypertensive therapies; and others, such as ischemic cardiomyopathy, have been reported with increased prevalence likely due to longer survival and higher rates of chronic disease. ADPKD-specific screening and management guidelines exist for hypertension, LVH, and ICAs; and these are described in this review.
Collapse
Affiliation(s)
- Priyanka S. Sagar
- Michael Stern Laboratory for Polycystic Kidney Disease, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
- Department of Renal Medicine, Nepean Hospital, Nepean Blue Mountains Local Health District, Sydney, New South Wales, Australia
| | - Gopala K. Rangan
- Michael Stern Laboratory for Polycystic Kidney Disease, Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Daniel EA, Sommer NA, Sharma M. Polycystic kidneys: interaction of notch and renin. Clin Sci (Lond) 2023; 137:1145-1150. [PMID: 37553961 PMCID: PMC11132639 DOI: 10.1042/cs20230023] [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: 05/28/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023]
Abstract
Polycystic kidney disease (PKD) is a developmental disorder, which either manifests in early childhood or later in life, depending on the genetic mutation one harbors. The mechanisms of cyst initiation are not well understood. Increasing literature is now suggesting that Notch signaling may play a critical role in PKD. Activation of Notch signaling is important during nephrogenesis and slows down after development. Deletion of various Notch molecules in the cap mesenchyme leads to formation of cysts and early death in mice. A new study by Belyea et al. has now found that cells of renin lineage may link Notch expression and cystic kidney disease. Here, we use our understanding of Notch signaling and PKD to speculate about the significance of these interactions.
Collapse
Affiliation(s)
- Emily A Daniel
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| | - Nicole A Sommer
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| | - Madhulika Sharma
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas 66160, U.S.A
| |
Collapse
|
5
|
Pana C, Stanigut AM, Cimpineanu B, Alexandru A, Salim C, Nicoara AD, Resit P, Tuta LA. Urinary Biomarkers in Monitoring the Progression and Treatment of Autosomal Dominant Polycystic Kidney Disease-The Promised Land? MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050915. [PMID: 37241147 DOI: 10.3390/medicina59050915] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease, and it leads to end-stage renal disease (ESRD). The clinical manifestations of ADPKD are variable, with extreme differences observable in its progression, even among members of the same family with the same genetic mutation. In an age of new therapeutic options, it is important to identify patients with rapidly progressive evolution and the risk factors involved in the disease's poor prognosis. As the pathophysiological mechanisms of the formation and growth of renal cysts have been clarified, new treatment options have been proposed to slow the progression to end-stage renal disease. Furthermore, in addition to the conventional factors (PKD1 mutation, hypertension, proteinuria, total kidney volume), increasing numbers of studies have recently identified new serum and urinary biomarkers of the disease's progression, which are cheaper and more easily to dosing from the early stages of the disease. The present review discusses the utility of new biomarkers in the monitoring of the progress of ADPKD and their roles in new therapeutic approaches.
Collapse
Affiliation(s)
- Camelia Pana
- Nephrology Department, Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Alina Mihaela Stanigut
- Nephrology Department, Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Bogdan Cimpineanu
- Medical Semiology Department, Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Andreea Alexandru
- Nephrology Department, Constanta County Emergency Hospital, 900601 Constanta, Romania
| | - Camer Salim
- Emergency Department, Constanta County Emergency Hospital, 900601 Constanta, Romania
| | - Alina Doina Nicoara
- Medical Semiology Department, Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| | - Periha Resit
- Faculty of Medicine, "Ovidius" University of Constanta, 900601 Constanta, Romania
| | - Liliana Ana Tuta
- Nephrology Department, Faculty of Medicine, "Ovidius" University of Constanta, 900470 Constanta, Romania
| |
Collapse
|
6
|
Lucchetti L, Chinali M, Emma F, Massella L. Autosomal dominant and autosomal recessive polycystic kidney disease: hypertension and secondary cardiovascular effect in children. Front Mol Biosci 2023; 10:1112727. [PMID: 37006611 PMCID: PMC10064450 DOI: 10.3389/fmolb.2023.1112727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Autosomal dominant (ADPKD) and autosomal recessive (ARPKD) polycystic kidney disease are the most widely known cystic kidney diseases. They are significantly different from each other in terms of genetics and clinical manifestations. Hypertension is one of the main symptoms in both diseases, but the age of onset and secondary cardiovascular complications are significantly different. Most ARPKD children are hypertensive in the first year of life and need high doses of hypertensive drugs. ADPKD patients with a very early onset of the disease (VEOADPKD) develop hypertension similarly to patients with ARPKD. Conversely, a significantly lower percentage of patients with classic forms of ADPKD develops hypertension during childhood, although probably more than originally thought. Data published in the past decades show that about 20%–30% of ADPKD children are hypertensive. Development of hypertension before 35 years of age is a known risk factor for more severe disease in adulthood. The consequences of hypertension on cardiac geometry and function are not well documented in ARPKD due to the rarity of the disease, the difficulties in collecting homogeneous data, and differences in the type of parameters evaluated in different studies. Overall, left ventricular hypertrophy (LVH) has been reported in 20%–30% of patients and does not always correlate with hypertension. Conversely, cardiac geometry and cardiac function are preserved in the vast majority of hypertensive ADPKD children, even in patients with faster decline of kidney function. This is probably related to delayed onset of hypertension in ADPKD, compared to ARPKD. Systematic screening of hypertension and monitoring secondary cardiovascular damage during childhood allows initiating and adapting antihypertensive treatment early in the course of the disease, and may limit disease burden later in adulthood.
Collapse
Affiliation(s)
- L. Lucchetti
- Division of Nephrology, Department of Paediatric Subspecialties, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - M. Chinali
- Department of Cardiac Surgery, Cardiology and Heart Lung Transplant, Bambino Gesù Children’s Hospital (IRCCS), Rome, Italy
| | - F. Emma
- Division of Nephrology, Department of Paediatric Subspecialties, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - L. Massella
- Division of Nephrology, Department of Paediatric Subspecialties, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: L. Massella,
| |
Collapse
|
7
|
Fountain HB, de Monaco BA, Jagid J, Benveniste R, Cordeiro JG. Middle Meningeal Artery Embolization for Chronic Subdural Hematoma Related to Autosomal Dominant Polycystic Kidney Disease. Cureus 2023; 15:e34970. [PMID: 36938171 PMCID: PMC10019495 DOI: 10.7759/cureus.34970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 02/16/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a connective tissue disease with vascular abnormalities involving multiple organs. The prevalence of ADPKD associated with a spontaneous subdural hematoma (SDH) is very low, with less than 10 cases reported in the literature to date. Symptomatic chronic SDH is classically treated with a twist drill, burr holes, or craniotomy. Recently, middle meningeal artery (MMA) embolization has emerged as an ancillary modality. We present the first case in the literature of a bilateral SDH in a young ADPKD patient successfully managed with MMA embolization. Moreover, we discuss the role of different treatment modalities on this subset of patients.
Collapse
Affiliation(s)
- Hayes B Fountain
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Bernardo A de Monaco
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
- Neurosurgery, University of Sao Paulo, Sao Paulo, BRA
| | - Jonathan Jagid
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Ronald Benveniste
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| | - Joacir G Cordeiro
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
8
|
Abstract
Circulating blood is filtered across the glomerular barrier to form an ultrafiltrate of plasma in the Bowman's space. The volume of glomerular filtration adjusted by time is defined as the glomerular filtration rate (GFR), and the total GFR is the sum of all single-nephron GFRs. Thus, when the single-nephron GFR is increased in the context of a normal number of functioning nephrons, single glomerular hyperfiltration results in 'absolute' hyperfiltration in the kidney. 'Absolute' hyperfiltration can occur in healthy people after high protein intake, during pregnancy and in patients with diabetes, obesity or autosomal-dominant polycystic kidney disease. When the number of functioning nephrons is reduced, single-nephron glomerular hyperfiltration can result in a GFR that is within or below the normal range. This 'relative' hyperfiltration can occur in patients with a congenitally reduced nephron number or with an acquired reduction in nephron mass consequent to surgery or kidney disease. Improved understanding of the mechanisms that underlie 'absolute' and 'relative' glomerular hyperfiltration in different clinical settings, and of whether and how the single-nephron haemodynamic and related biomechanical forces that underlie glomerular hyperfiltration promote glomerular injury, will pave the way toward the development of novel therapeutic interventions that attenuate glomerular hyperfiltration and potentially prevent or limit consequent progressive kidney injury and loss of function.
Collapse
|
9
|
Park HC, Oh YK. Practical Issues in the Management of Polycystic Kidney Disease: Blood Pressure and Water Balance. Electrolyte Blood Press 2022; 20:10-16. [PMID: 36451711 PMCID: PMC9685325 DOI: 10.5049/ebp.2022.20.1.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/06/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Autosomal dominant polycystic kidney disease is the most common hereditary renal disease affecting more than 13 million people worldwide. Renal function deteriorates as the cysts in both kidneys increase in number and size, which eventually results in end-stage kidney failure. Until recently, conservative management for chronic kidney disease such as blood pressure control, low sodium diet, adequate water intake, and weight control were known for the only treatment of polycystic kidney disease. However, the introduction of disease-modifying drug has led to the new paradigm shift in the management of polycystic kidney disease. Tolvaptan, the vasopressin V2 receptor antagonist, has been introduced to the patients with large kidneys since it can inhibit cyclic adenosine monophosphate, attenuates cyst growth, and delays renal failure. This article reviews the two important practical issues in the management of polycystic kidney disease: blood pressure and water balance. Firstly, the article will review the pathogenesis of high blood pressure in polycystic kidney disease and will demonstrate the current up-to-date management plan for blood pressure control. Secondly, this article will explain the mechanism of Tolvaptan on the treatment of polycystic kidney disease and its possible adverse effect on water and sodium balance.
Collapse
Affiliation(s)
- Hayne Cho Park
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Department of Internal Medicine, Kangnam Sacred Heart Hospital, Seoul, Republic of Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | | |
Collapse
|
10
|
Blijdorp CJ, Severs D, Musterd-Bhaggoe UM, Gansevoort RT, Zietse R, Hoorn EJ. Serum bicarbonate is associated with kidney outcomes in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant 2021; 36:2248-2255. [PMID: 33377160 PMCID: PMC8643593 DOI: 10.1093/ndt/gfaa283] [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] [Received: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Metabolic acidosis accelerates progression of chronic kidney disease, but whether this is also true for autosomal dominant polycystic kidney disease (ADPKD) is unknown. METHODS Patients with ADPKD from the DIPAK (Developing Interventions to halt Progression of ADPKD) trial were included [n = 296, estimated glomerular filtration rate (eGFR) 50 ± 11 mL/min/1.73 m2, 2.5 years follow-up]. Outcomes were worsening kidney function (30% decrease in eGFR or kidney failure), annual eGFR change and height-adjusted total kidney and liver volumes (htTKV and htTLV). Cox and linear regressions were adjusted for prognostic markers for ADPKD [Mayo image class and predicting renal outcomes in ADPKD (PROPKD) scores] and acid-base parameters (urinary ammonium excretion). RESULTS Patients in the lowest tertile of baseline serum bicarbonate (23.1 ± 1.6 mmol/L) had a significantly greater risk of worsening kidney function [hazard ratio = 2.95, 95% confidence interval (CI) 1.21-7.19] compared with patients in the highest tertile (serum bicarbonate 29.0 ± 1.3 mmol/L). Each mmol/L decrease in serum bicarbonate increased the risk of worsening kidney function by 21% in the fully adjusted model (hazard ratio = 1.21, 95% CI 1.06-1.37). Each mmol/L decrease of serum bicarbonate was also associated with further eGFR decline (-0.12 mL/min/1.73 m2/year, 95% CI -0.20 to -0.03). Serum bicarbonate was not associated with changes in htTKV or htTLV growth. CONCLUSIONS In patients with ADPKD, a lower serum bicarbonate within the normal range predicts worse kidney outcomes independent of established prognostic factors for ADPKD and independent of urine ammonium excretion. Serum bicarbonate may add to prognostic models and should be explored as a treatment target in ADPKD.
Collapse
Affiliation(s)
- Charles J Blijdorp
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - David Severs
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Usha M Musterd-Bhaggoe
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ronald T Gansevoort
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert Zietse
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
11
|
Li Q, Wang Y, Deng W, Liu Y, Geng J, Yan Z, Li F, Chen B, Li Z, Xia R, Zeng W, Liu R, Xu J, Xiong F, Wu CL, Miao Y. Heterogeneity of cell composition and origin identified by single-cell transcriptomics in renal cysts of patients with autosomal dominant polycystic kidney disease. Theranostics 2021; 11:10064-10073. [PMID: 34815804 PMCID: PMC8581434 DOI: 10.7150/thno.57220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Rationale: Renal cysts in patients with autosomal dominant polycystic kidney disease (ADPKD) can originate from any nephron segments, including proximal tubules (PT), the loop of Henle (LOH), distal tubules (DT), and collecting ducts (CD). Previous studies mostly used limited cell markers and failed to identify cells negative for these markers. Therefore, the cell composition and origin of ADPKD cyst are still unclear, and mechanisms of cystogenesis of different origins await further exploration. Methods: We performed single-cell RNA sequencing for the normal kidney tissue and seven cysts derived from superficial or deep layers of the polycystic kidney from an ADPKD patient. Results: Twelve cell types were identified and analyzed. We found that a renal cyst could be derived either from CD or both PT and LOH. Gene set variation analysis (GSVA) showed that epithelial mesenchymal transition (EMT), TNFA signaling via the NFKB pathways, and xenobiotic metabolism were significantly activated in PT-derived cyst epithelial cells while robust expression of genes involved in G2M Checkpoint, mTORC1 signaling, E2F Targets, MYC Targets V1, MYC Targets V2 were observed in CD-derived cells. Conclusion: Our results revealed that a single cyst could originate from CD or both PT and LOH, suggesting heterogeneity of polycystic composition and origin. Furthermore, cyst epithelial cells with different origins have different gene set activation.
Collapse
|
12
|
Vasileva VY, Sultanova RF, Sudarikova AV, Ilatovskaya DV. Insights Into the Molecular Mechanisms of Polycystic Kidney Diseases. Front Physiol 2021; 12:693130. [PMID: 34566674 PMCID: PMC8456103 DOI: 10.3389/fphys.2021.693130] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant (AD) and autosomal recessive (AR) polycystic kidney diseases (PKD) are severe multisystem genetic disorders characterized with formation and uncontrolled growth of fluid-filled cysts in the kidney, the spread of which eventually leads to the loss of renal function. Currently, there are no treatments for ARPKD, and tolvaptan is the only FDA-approved drug that alleviates the symptoms of ADPKD. However, tolvaptan has only a modest effect on disease progression, and its long-term use is associated with many side effects. Therefore, there is still a pressing need to better understand the fundamental mechanisms behind PKD development. This review highlights current knowledge about the fundamental aspects of PKD development (with a focus on ADPKD) including the PC1/PC2 pathways and cilia-associated mechanisms, major molecular cascades related to metabolism, mitochondrial bioenergetics, and systemic responses (hormonal status, levels of growth factors, immune system, and microbiome) that affect its progression. In addition, we discuss new information regarding non-pharmacological therapies, such as dietary restrictions, which can potentially alleviate PKD.
Collapse
Affiliation(s)
| | - Regina F Sultanova
- Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia.,Department of Physiology, Augusta University, Augusta, GA, United States
| | | | | |
Collapse
|
13
|
Gul CB, Yildiz A, Sag S, Oruc A, Ersoy A, Gullulu S. The Effect of Smoking on Endothelial Dysfunction in Autosomal Dominant Polycystic Kidney Disease Patients with Preserved Renal Function. Ren Fail 2021; 43:1124-1129. [PMID: 34256663 PMCID: PMC8279153 DOI: 10.1080/0886022x.2021.1949348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background In autosomal dominant polycystic kidney disease (ADPKD), endothelial dysfunction (ED) is common and occurs much earlier than kidney function impairment. The impact of smoking on ED in ADPKD patients has not been previously studied. The aim of this study was to investigate the potential contribution of smoking habits to ED and subclinical atherosclerosis in these patients. Methods This case-control study included 54 ADPKD patients with preserved renal function and 45 healthy control subjects. ED was assessed using ischemia-induced forearm flow-mediated dilatation (FMD). Carotid intima-media thickness (CIMT) was measured from 10 mm proximal to the right common carotid artery. Clinical demographic characteristics and laboratory data were recorded for the patients and control group. Regression analysis was used to determine independent associations of ED and CIMT. Results FMD was significantly lower in the ADPKD patients (19.5 ± 5.63 vs. 16.56 ± 6.41, p = .018). Compared with nonsmoker ADPKD patients, smoker patients had significantly lower FMD values (18.19 ± 6.52 vs. 13.79 ± 5.27, p = .013). In multiple regression analysis, age (β = –0.294, 95% CI: −0.392: −1.96, p = .001) for FMD and smoking (β = 1.328, 95% CI: 0.251, 2.404, p = .017) for CIMT were independent predictors. Conclusions Patients with ADPKD had more impaired endothelial function and subclinical atherosclerosis compared with control subjects. Smoking may increase the risk of subclinical atherosclerosis in ADPKD patients.
Collapse
Affiliation(s)
- Cuma Bulent Gul
- Department of Nephrology, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Abdulmecit Yildiz
- Department of Nephrology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Saim Sag
- Department of Cardiology, Acibadem University Faculty of Medicine, Istanbul, Turkey
| | - Aysegul Oruc
- Department of Nephrology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Alparslan Ersoy
- Department of Nephrology, Uludag University Faculty of Medicine, Bursa, Turkey
| | - Sumeyye Gullulu
- Department of Cardiology, Uludag University Faculty of Medicine, Bursa, Turkey
| |
Collapse
|
14
|
Mahendran R, Lim SK, Ong KC, Chua KH, Chai HC. Natural-derived compounds and their mechanisms in potential autosomal dominant polycystic kidney disease (ADPKD) treatment. Clin Exp Nephrol 2021; 25:1163-1172. [PMID: 34254206 DOI: 10.1007/s10157-021-02111-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/06/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic kidney disorder that impairs renal functions progressively leading to kidney failure. The disease affects between 1:400 and 1:1000 ratio of the people worldwide. It is caused by the mutated PKD1 and PKD2 genes which encode for the defective polycystins. Polycystins mimic the receptor protein or protein channel and mediate aberrant cell signaling that causes cystic development in the renal parenchyma. The cystic development is driven by the increased cyclic AMP stimulating fluid secretion and infinite cell growth. In recent years, natural product-derived small molecules or drugs targeting specific signaling pathways have caught attention in the drug discovery discipline. The advantages of natural products over synthetic drugs enthusiast researchers to utilize the medicinal benefits in various diseases including ADPKD. CONCLUSION Overall, this review discusses some of the previously studied and reported natural products and their mechanisms of action which may potentially be redirected into ADPKD.
Collapse
Affiliation(s)
- Rhubaniya Mahendran
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Soo Kun Lim
- Renal Division, Department of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Hwa Chia Chai
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
15
|
Li S, Hildreth CM, Rahman AA, Barton SA, Wyse BF, Lim CK, Pilowsky PM, Phillips JK. Renal denervation does not affect hypertension or the renin-angiotensin system in a rodent model of juvenile-onset polycystic kidney disease: clinical implications. Sci Rep 2021; 11:14286. [PMID: 34253766 PMCID: PMC8275789 DOI: 10.1038/s41598-021-93575-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
We examined the effect of total and afferent renal denervation (RDN) on hypertension and the renin-angiotensin system (RAS) in a rodent model of juvenile-onset polycystic kidney disease (PKD). Lewis Polycystic Kidney (LPK) and control rats received total, afferent or sham RDN by periaxonal application of phenol, capsaicin or normal saline, respectively, and were monitored for 4-weeks. Afferent RDN did not affect systolic blood pressure (SBP) determined by radiotelemetry in either strain (n = 19) while total RDN significantly reduced SBP in Lewis rats 4-weeks post-denervation (total vs. sham, 122 ± 1 vs. 130 ± 2 mmHg, P = 0.002, n = 25). Plasma and kidney renin content determined by radioimmunoassay were significantly lower in LPK vs. Lewis (plasma: 278.2 ± 6.7 vs. 376.5 ± 11.9 ng Ang I/ml/h; kidney: 260.1 ± 6.3 vs. 753.2 ± 37.9 ng Ang I/mg/h, P < 0.001, n = 26). These parameters were not affected by RDN. Intrarenal mRNA expression levels of renin, angiotensinogen, angiotensin-converting enzyme (ACE)2, and angiotensin II receptor type 1a were significantly lower, whereas ACE1 expression was significantly higher in the LPK vs. Lewis (all P < 0.05, n = 26). This pattern of intrarenal RAS expression was not changed by RDN. In conclusion, RDN does not affect hypertension or the RAS in the LPK model and indicates RDN might not be a suitable antihypertensive strategy for individuals with juvenile-onset PKD.
Collapse
Affiliation(s)
- Sheran Li
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Cara M Hildreth
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Ahmed A Rahman
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Sean A Barton
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Benjamin F Wyse
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Chai K Lim
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Paul M Pilowsky
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jacqueline K Phillips
- Department of Biomedical Sciences, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
16
|
Lu CL, Lin CY, Lin LY, Chen PC, Zheng CM, Lu KC, Yeih DF. Primary prevention of cardiovascular disease events with renin-angiotensin system blockade in autosomal dominant polycystic kidney disease dialysis patients: A nationwide cohort study. Medicine (Baltimore) 2021; 100:e26559. [PMID: 34190195 PMCID: PMC8257834 DOI: 10.1097/md.0000000000026559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 06/12/2021] [Indexed: 01/04/2023] Open
Abstract
Although renin-angiotensin system (RAS) blockade has been shown to reduce cardiovascular disease (CVD) in the general population and high-risk subjects, their protective effect in autosomal dominant polycystic kidney disease (ADPKD) patients under dialysis was still unknown. By using the database from 1995 to 2008 Taiwan National Health Insurance Research Database (Registry for Catastrophic Illnesses), we included 387 ADPKD patients who received dialysis therapy, aged ≥ 18 year-old, and with no evidence of CVD events in 1997 and 1998. We utilized Cox proportional hazards regression analysis and propensity score matching to evaluate adjusted hazard ratios for all-cause mortality and CVD events in users (n=231) and nonusers (n = 156) of an angiotensin-converting enzymes inhibitor (ACEI) / angiotensin II receptor blocker (ARB) during the 12 years of follow-up. All study subjects were followed up for more than 3 months. There was no significant difference between the ACEI/ARB treatment group and the control group in incident CVD events except ischemic stroke and transient ischemic accident (TIA). The results remain similar between groups before and after propensity score matching. Moreover, there was no significant difference in outcomes between ACEI/ARB treatment over 50% of follow-up period and without ACEI/ARB treatment after propensity score matching. This nationwide cohort study failed to prove the protective effects of long-term ACEI or ARB on incident CVD events among APKD dialysis patients. Further larger scale, multicenter and randomized control trials are warranted to show the causal association.
Collapse
Affiliation(s)
- Chien-Lin Lu
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital
- School of Medicine, College of Medicine, Fu Jen Catholic University
| | - Chien-Yu Lin
- School of Medicine, College of Medicine, Fu Jen Catholic University
- Division of Nephrology, Department of Internal Medicine, En Chu Kong Hospital
- Department of Environmental Engineering and Health, Yuanpei University of Medical Technology, Hsinchu
| | - Lian-Yu Lin
- Division of Cardiology, Department of Medicine, National Taiwan University Hospital
| | - Pau-Chung Chen
- Institute of Environmental and Occupational Health Sciences, National Taiwan University College of Public Health
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital
- Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation
| | - Dong-Feng Yeih
- School of Medicine, College of Medicine, Fu Jen Catholic University
- Division of Cardiology, Department of Medicine, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| |
Collapse
|
17
|
Song R, Yosypiv IV. Sequence variants in the renin-angiotensin system genes are associated with isolated multicystic dysplastic kidney in children. Pediatr Res 2021; 90:205-211. [PMID: 33173183 DOI: 10.1038/s41390-020-01255-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/06/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Multicystic dysplastic kidney (MCDK) is a common form of congenital cystic kidney disease in children. The etiology of MCDK remains unclear. Given an important role of the renin-angiotensin system in normal kidney development, we explored whether MCDK in children is associated with variants in the genes encoding renin-angiotensin system components by Sanger sequencing. METHODS The coding regions of renin (REN), angiotensinogen (AGT), ACE, and angiotensin 1 receptor (AGTR1) genes were amplified by PCR. The effect of DNA sequence variants on protein function was predicted with PolyPhen-2 software. RESULTS 3 novel and known AGT variants were found. 1 variant was probably damaging, 1 was possibly damaging and one was benign. Out of 7 REN variants, 4 were probably damaging and 3 were benign. Of 6 ACE variants, 3 were probably damaging and 3-benign. 3 AGTR1 variants were found. 2 variants were possibly damaging, and one was benign. CONCLUSION We report novel associations of sequence variants in REN, AGT, ACE, or AGTR1 genes in children with isolated MCDK in the United States. Our findings suggest a recessive disease model and support the hypothesis of multiple renin-angiotensin system gene involvement in MCDK. IMPACT Discovery of novel gene variants in renin-angiotensin genes in children with MCDK. Novel possibly damaging gene variants discovered. Multiple renin-angiotensin system gene variants are involved in MCDK.
Collapse
Affiliation(s)
- Renfang Song
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Ihor V Yosypiv
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
18
|
Leierer J, Perco P, Hofer B, Eder S, Dzien A, Kerschbaum J, Rudnicki M, Mayer G. Coregulation Analysis of Mechanistic Biomarkers in Autosomal Dominant Polycystic Kidney Disease. Int J Mol Sci 2021; 22:6885. [PMID: 34206927 PMCID: PMC8269435 DOI: 10.3390/ijms22136885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder leading to deterioration of kidney function and end stage kidney disease (ESKD). A number of molecular processes are dysregulated in ADPKD but the exact mechanism of disease progression is not fully understood. We measured protein biomarkers being linked to ADPKD-associated molecular processes via ELISA in urine and serum in a cohort of ADPKD patients as well as age, gender and eGFR matched CKD patients and healthy controls. ANOVA and t-tests were used to determine differences between cohorts. Spearman correlation coefficient analysis was performed to assess coregulation patterns of individual biomarkers and renal function. Urinary epidermal growth factor (EGF) and serum apelin (APLN) levels were significantly downregulated in ADPKD patients. Serum vascular endothelial growth factor alpha (VEGFA) and urinary angiotensinogen (AGT) were significantly upregulated in ADPKD patients as compared with healthy controls. Arginine vasopressin (AVP) was significantly upregulated in ADPKD patients as compared with CKD patients. Serum VEGFA and VIM concentrations were positively correlated and urinary EGF levels were negatively correlated with urinary AGT levels. Urinary EGF and AGT levels were furthermore significantly associated with estimated glomerular filtration rate (eGFR) in ADPKD patients. In summary, altered protein concentrations in body fluids of ADPKD patients were found for the mechanistic markers EGF, APLN, VEGFA, AGT, AVP, and VIM. In particular, the connection between EGF and AGT during progression of ADPKD warrants further investigation.
Collapse
Affiliation(s)
- Johannes Leierer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Paul Perco
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Benedikt Hofer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Susanne Eder
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | | | - Julia Kerschbaum
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Michael Rudnicki
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| |
Collapse
|
19
|
Pugliese NR, Masi S, Taddei S. The renin-angiotensin-aldosterone system: a crossroad from arterial hypertension to heart failure. Heart Fail Rev 2020; 25:31-42. [PMID: 31512149 DOI: 10.1007/s10741-019-09855-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin-aldosterone system (RAAS) plays a pivotal role in the regulation of blood pressure and volume homeostasis, promoting critical structural changes in every component of the cardiovascular system, including the heart and blood vessels. Consequently, the RAAS is a crucial therapeutic target for several chronic diseases of the cardiovascular system, spanning from arterial hypertension (AH) to heart failure (HF). AH represents a leading risk factor for the development of symptomatic HF, particularly with left ventricle (LV) preserved ejection fraction (HFpEF). LV diastolic dysfunction and cardiac remodelling are the first discernible manifestations of heart disease in patients with AH. Typically, AH develops many years before the diagnosis of overt HF, providing a therapeutic target for preventive strategies. Treatment of AH is based on different classes of antihypertensive drugs, which show differences in their capacity to prevent the evolution towards HF. The blockers of the RAAS are effective drugs to treat AH and prevent HF with reduced ejection fraction (HFrEF), but the evidence of the potential benefits in patients with HFpEF remains limited. In this review, the authors summarise data from several clinical trials of HFpEF and HFrEF, focusing on the mechanisms leading the transition from AH to HF and late complications.
Collapse
Affiliation(s)
- Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy.
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma, 67, 56126, Pisa, Italy
| |
Collapse
|
20
|
Lai S, Mangiulli M, Perrotta AM, Di Lazzaro Giraldi G, Testorio M, Rosato E, Cianci R, Gigante A. Reduction in Heart Rate Variability in Autosomal Dominant Polycystic Kidney Disease. Kidney Blood Press Res 2019; 44:1142-1148. [PMID: 31550720 DOI: 10.1159/000502419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 07/30/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Cardiovascular disease is one of the main causes of morbidity and mortality in patients with autosomal dominant polycystic kidney disease (ADPKD). Autonomic dysfunction is associated with an increased risk for all cardiovascular events in the general population and can be evaluated with heart rate variability (HRV). OBJECTIVE To evaluate HRV in ADPKD patients with mild hypertension versus hypertensive patients with organ damage and healthy controls (HC). MATERIALS AND METHODS We have enrolled 65 patients: 21 ADPKD patients (10 males), 20 patients with hypertension (14 males), and 24 HC (10 males). Biochemical analysis, clinical evaluation, anthropometric data, intima-media thickness, 24-h ECG Holter recording, and echocardiography were investigated at the time of enrollment. RESULTS No significant differences in HRV parameters were found between ADPKD with mild hypertension and hypertensive patients with organ damage. The median of HRV variables in time domain as SDNN (global autonomic activity) was significantly lower in ADPKD and hypertensive patients than HC (p < 0.05). In the frequency domain analysis, low frequency (LF), which mainly reflects the sympathetic component, showed higher values in ADPKD and hypertensive patients than HC during the night (p < 0.01). During the night, the sympathovagal balance, LF/high frequency (HF), showed higher values in ADPKD and hypertensive patients than HC (p < 0.0001). Conversely, LF day was lower in ADPKD and hypertensive patients than HC (p < 0.01). HF, which mainly reflects the parasympathetic component, was lower in ADPKD and hypertensive patients during the night than HC (p < 0.0001). CONCLUSIONS HRV reduction is present in ADPKD patients with mild hypertension in the absence of organ damage. The evaluation of sympathovagal balance can provide novel information on the cardiovascular risk in ADPKD patients in addition to classical risk factors.
Collapse
Affiliation(s)
- Silvia Lai
- Nephrology Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy,
| | - Marco Mangiulli
- Nephrology Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Adolfo M Perrotta
- Nephrology Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Massimo Testorio
- Nephrology Unit, Department of Gynecologic-Obstetrical Sciences and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario Cianci
- Nephrology Unit, Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
21
|
Brosnahan GM, Abebe KZ, Moore CG, Bae KT, Braun WE, Chapman AB, Flessner MF, Harris PC, Hogan MC, Perrone RD, Rahbari-Oskoui FF, Steinman TI, Torres VE, The Halt Pkd Investigators. Determinants of Progression in Early Autosomal Dominant Polycystic Kidney Disease: Is it Blood Pressure or Renin-Angiotensin-Aldosterone-System Blockade? Curr Hypertens Rev 2019; 14:39-47. [PMID: 29564978 PMCID: PMC6063360 DOI: 10.2174/1573402114666180322110209] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 12/17/2022]
Abstract
Background The HALT PKD trial in early autosomal dominant polycystic kidney disease (ADPKD) showed that intensive control of systolic blood pressure to 95-110 mmHg was associated with a 14% slower rate of kidney volume growth compared to standard control. It is unclear whether this result was due to greater blockade of the renin-angiotensin-aldosterone system (RAAS) by allowing the use of higher drug doses in the low blood pressure arm, or due to the lower blood pressure per se. Methods In this secondary analysis of HALT PKD Study A, we categorized participants into high and low dose groups based on the median daily equivalent dose of RAAS blocking drugs used after the initial dose titration period. Using linear mixed models, we compared the percent change in total kidney volume and the slope of estimated glomerular filtration rate (eGFR) between the 2 groups. We also assessed the effects of time-varying dose and time-varying blood pressure parameters on these outcomes. Results Subjects in the high dose group (n=252) did not experience a slower increase in total kidney volume than those in the low-dose (n=225) group, after adjustment for age, sex, genotype, and BP arm. The chronic slope of eGFR decline was similar in the 2 groups. Higher time-varying systolic blood pressure was associated with a steeper decline in eGFR. Conclusion ADPKD progression (as detected by eGFR decline and TKV increase) was ameliorated by intense blood pressure control as opposed to pharmacologic intensity of RAAS blockade.
Collapse
Affiliation(s)
| | - Kaleab Z Abebe
- University of Pittsburgh, Pittsburgh, Pennsylvania, PA, United States
| | - Charity G Moore
- University of Pittsburgh, Pittsburgh, Pennsylvania, PA, United States
| | - Kyongtae T Bae
- University of Pittsburgh, Pittsburgh, Pennsylvania, PA, United States
| | | | | | | | | | - Marie C Hogan
- Mayo Clinic, Rochester, Minnesota, MN, United States
| | | | | | - Theodore I Steinman
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, MA, United States
| | | | | |
Collapse
|
22
|
Kim H, Park S, Jhee JH, Yun HR, Park JT, Han SH, Lee J, Kim SW, Kim YH, Oh YK, Kang SW, Choi KH, Yoo TH. Urinary angiotensinogen level is associated with potassium homeostasis and clinical outcome in patients with polycystic kidney disease: a prospective cohort study. BMC Nephrol 2019; 20:104. [PMID: 30909873 PMCID: PMC6434770 DOI: 10.1186/s12882-019-1292-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 03/11/2019] [Indexed: 01/13/2023] Open
Abstract
Background Guidelines for general hypertension treatment do not recommend the combined use of renin-angiotensin-aldosterone system (RAAS) inhibitors due to the risk of hyperkalemia. However, a recent clinical trial showed that polycystic kidney disease (PKD) patients had infrequent episodes of hyperkalemia despite receiving combined RAAS inhibitors. Because intrarenal RAAS is a main component for renal potassium handling, we further investigated the association between intrarenal RAAS activity and serum potassium level in patients with chronic kidney disease, particularly in PKD patients, and examined whether intrarenal RAAS activity has a prognostic role in patients with PKD. Methods A total of 1788 subjects from the KoreaN cohort study for Outcome in patients With Chronic Kidney Disease (KNOW-CKD) were enrolled in this study. Intrarenal RAAS activity was assessed by the measurement of urinary angiotensinogen (AGT). The primary outcome was the composite of all-cause mortality and renal function decline. Results Patients with PKD had a significantly lower serum potassium level in chronic kidney disease stages 1 to 3b than non-PKD patients. In logistic regression analysis, after adjusting for multiple confounders, PKD patients had a significantly lower risk of hyperkalemia than non-PKD patients. In multivariable linear regression analysis, the urinary AGT/creatinine (Cr) ratio was negatively correlated with the serum potassium level (β = − 0.058, P = 0.017) and positively correlated with the transtubular potassium gradient (TTKG, β = 0.087, P = 0.001). In propensity score matching analysis, after matching factors associated with serum potassium and TTKG, PKD patients had a significantly higher TTKG (P = 0.021) despite a lower serum potassium level (P = 0.004). Additionally, the urinary AGT/Cr ratio was significantly higher in PKD patients than in non-PKD patients (P = 0.011). In 293 patients with PKD, high urinary AGT/Cr ratio was associated with increased risk of the composite outcome (hazard ratio 1.29; 95% confidence interval, 1.07–1.55; P = 0.007). Conclusions High activity of intrarenal RAAS is associated with increased urinary potassium excretion and low serum potassium level in patients with PKD. In addition, intrarenal RAAS activity can be a prognostic marker for mortality and renal function decline in these patients.
Collapse
Affiliation(s)
- Hyoungnae Kim
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea.,Division of Nephrology, Soonchunhyang University Hospital, Seoul, Republic of Korea
| | - Seohyun Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jong Hyun Jhee
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University College of Medicine, Incheon, Republic of Korea
| | - Hae-Ryong Yun
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jung Tak Park
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Hyeok Han
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Joongyub Lee
- Medical Research Collaborating Center, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Busan Paik Hospital, Inje University, Busan, Republic of Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center, Seoul, Republic of Korea
| | - Shin-Wook Kang
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Kyu Hun Choi
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea
| | - Tae-Hyun Yoo
- Department of Internal Medicine, College of Medicine, Institute of Kidney Disease Research, Yonsei University, Seoul, 03722, Republic of Korea.
| | | |
Collapse
|
23
|
Antisense oligonucleotides targeting angiotensinogen: insights from animal studies. Biosci Rep 2019; 39:BSR20180201. [PMID: 30530571 PMCID: PMC6328882 DOI: 10.1042/bsr20180201] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/21/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023] Open
Abstract
Angiotensinogen (AGT) is the unique substrate of all angiotensin peptides. We review the recent preclinical research of AGT antisense oligonucleotides (ASOs), a rapidly evolving therapeutic approach. The scope of the research findings not only opens doors for potentially new therapeutics of hypertension and many other diseases, but also provides insights into understanding critical physiological and pathophysiological roles mediated by AGT.
Collapse
|
24
|
Lai S, Mastroluca D, Letizia C, Petramala L, Perrotta AM, DiGaeta A, Ferrigno L, Ciccariello M, D'Angelo AR, Panebianco V. Magnetic resonance imaging 3T and total fibrotic volume in autosomal dominant polycystic kidney disease. Intern Med J 2018; 48:1505-1513. [DOI: 10.1111/imj.14039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 07/01/2018] [Accepted: 07/05/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Silvia Lai
- Department of Clinical Medicine; Sapienza University of Rome; Rome Italy
| | - Daniela Mastroluca
- Department of Nephrology and Dialysis Unit, Hospital ICOT Latina; Sapienza University of Rome; Rome Italy
| | - Claudio Letizia
- Department of Internal Medicine and Medical Specialities; Sapienza University of Rome; Rome Italy
| | - Luigi Petramala
- Department of Internal Medicine and Medical Specialities; Sapienza University of Rome; Rome Italy
| | - Adolfo M. Perrotta
- Department of Clinical Medicine; Sapienza University of Rome; Rome Italy
| | - Alessandro DiGaeta
- Department of Radiological, Oncological and Pathological Sciences; Sapienza University of Rome; Rome Italy
| | - Luigina Ferrigno
- National Centre for Epidemiology, Surveillance and Health Promotion; National Institute of Health; Rome Italy
| | - Mauro Ciccariello
- Department of Radiological, Oncological and Pathological Sciences; Sapienza University of Rome; Rome Italy
| | - Anna R. D'Angelo
- Department of Obstetrical-Gynecological Sciences and Urologic Sciences; Sapienza University of Rome; Rome Italy
| | - Valeria Panebianco
- Department of Radiological, Oncological and Pathological Sciences; Sapienza University of Rome; Rome Italy
| |
Collapse
|
25
|
Raptis V, Loutradis C, Sarafidis PA. Renal injury progression in autosomal dominant polycystic kidney disease: a look beyond the cysts. Nephrol Dial Transplant 2018; 33:1887-1895. [DOI: 10.1093/ndt/gfy023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Vasileios Raptis
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA Hospital, Thessaloniki, Greece
| | - Charalampos Loutradis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pantelis A Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
26
|
Lai S, Mastroluca D, Matino S, Panebianco V, Vitarelli A, Capotosto L, Turinese I, Marinelli P, Rossetti M, Galani A, Baiocchi P, D'Angelo AR, Palange P. Early Markers of Cardiovascular Risk in Autosomal Dominant Polycystic Kidney Disease. Kidney Blood Press Res 2017; 42:1290-1302. [PMID: 29262409 DOI: 10.1159/000486011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 06/06/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Cardiovascular disease is the most frequent cause of morbidity and mortality in autosomal dominant polycystic kidney disease (ADPKD) patients, often before the onset of renal failure, and the pathogenetic mechanism is not yet well elucidated. The aim of the study was to identify early and noninvasive markers of cardiovascular risk in young ADPKD patients, in the early stages of disease. METHODS A total of 26 patients with ADPKD and 24 control group, matched for age and sex, were enrolled, and we have assessed inflammatory indexes, mineral metabolism, metabolic state and markers of atherosclerosis and endothelial dysfunction (carotid intima media thickness (IMT), ankle brachial index (ABI), flow mediated dilation (FMD), renal resistive index (RRI), left ventricular mass index (LVMI)) and cardiopulmonary exercise testing (CPET), maximal O2 uptake (V'O2max), and O2 uptake at lactic acid threshold (V'O2@LT). RESULTS The ADPKD patients compared to control group, showed a significant higher mean value of LVMI, RRI, homocysteine (Hcy), Homeostasis Model Assessment-insulin resistance (HOMA-IR), serum uric acid (SUA), Cardiac-troponinT (cTnT) and intact parathyroid hormone (iPTH) (p<0.001, p<0.001, p<0.001, p<0.001, p<0.001, p=0.007, p=0.019; respectively), and a lower value of FMD and 25-hydroxyvitaminD (25-OH-VitD) (p<0.001, p<0.001) with reduced parameters of exercise tolerance, as V'O2max, V'O2max/Kg and V'O2max (% predicted) (p<0.001, p<0.001, p=0.018; respectively), and metabolic response indexes (V'O2@LT, V'O2 @LT%, V'O2@LT/Kg,) (p<0.001, p=0.14, p<0.001; respectively). Moreover, inflammatory indexes were significantly higher in ADPKD patients, and we found a positive correlation between HOMA-IR and C-reactive protein (CRP) (r=0.507, p=0.008), and a negative correlation between HOMA-IR and 25-OH-VitD (r=-0.585, p=0.002). CONCLUSION In our study, ADPKD patients, in the early stages of disease, showed a greater insulin resistance, endothelial dysfunction, inflammation and mineral metabolism disorders, respect to control group. Moreover, these patients presented reduced tolerance to stress, and decreased anaerobic threshold to CPET. Our results indicate a major and early cardiovascular risk in ADPKD patients. Therefore early and noninvasive markers of cardiovascular risk and CPET should be carried out, in ADPKD patients, in the early stages of disease, despite the cost implication.
Collapse
Affiliation(s)
- Silvia Lai
- Department of Clinical Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Mastroluca
- Nephrology and Dialysis Unit, Hospital ICOT Latina, Sapienza University of Rome, Rome, Italy
| | - Silvia Matino
- Nephrology, Dialysis and Trasplantation Unit, University of Bari, Bari, Italy
| | - Valeria Panebianco
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonio Vitarelli
- Department of Cardiovascular, Respiratory, Nephrological and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Lidia Capotosto
- Department of Cardiovascular, Respiratory, Nephrological and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Irene Turinese
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Paolo Marinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Marco Rossetti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Alessandro Galani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Pia Baiocchi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna R D'Angelo
- Department of Obstetrical-Gynecological Sciences and Urologic Sciences, Sapienza University of Rome, Rome, Italy
| | - Paolo Palange
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
27
|
Saito T, Tanaka Y, Morishita Y, Ishibashi K. Proteomic analysis of AQP11-null kidney: Proximal tubular type polycystic kidney disease. Biochem Biophys Rep 2017; 13:17-21. [PMID: 29204517 PMCID: PMC5709289 DOI: 10.1016/j.bbrep.2017.11.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 12/28/2022] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is caused by the mutation of polycystins (PC-1 or PC-2), in which cysts start from the collecting duct to extend to all nephron segments with eventual end stage renal failure. The cyst development is attenuated by a vasopressin V2 receptor antagonist tolvaptan which, however, will not affect proximal tubule cysts devoid of V2 receptor. Aquaporin-11 (AQP11) is expressed selectively in the proximal tubule of the kidney and AQP11-null kidneys have a disruptive PC-1 trafficking to the plasma membrane to develop polycystic kidneys. Here, we analyzed AQP11-null kidneys at the beginning of cyst formation by quantitative proteomic analysis using Tandem Mass Tag (TMT). Among ~ 1200 identified proteins, 124 proteins were differently expressed by > 1.5 or < 0.8 fold change. A pancreatic stone inhibitor or a growth factor, lithostathine-1 (Reg1) was most enhanced by 5 folds which was confirmed by western blot, while mitochondria-related proteins were downregulated. The identified proteins will be new target molecules for the treatment of proximal tubular cysts and helpful to explore the functional roles of AQP11 in the kidney. Proteomic analysis of the kidney from AQP11-null mice of proximal tubular specific ADPKD. Among ~ 1200 identified proteins, 124 proteins were differently expressed by > 1.5 or < 0.8 fold change. Mitochondrial proteins were downregulated reflecting a functional mitochondrial damage in cystic epithelia. Reg1 protein was most enhanced by 5 folds which was confirmed by western blot.
Collapse
Affiliation(s)
- Tatsuya Saito
- Department of Pathophysiology, Faculty of Clinical Pharmacy, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
- Corresponding author.
| | - Yasuko Tanaka
- Department of Pathophysiology, Faculty of Clinical Pharmacy, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Yoshiyuki Morishita
- Department of Nephrology, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Kenichi Ishibashi
- Department of Pathophysiology, Faculty of Clinical Pharmacy, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| |
Collapse
|
28
|
Fitzgibbon WR, Dang Y, Bunni MA, Baicu CF, Zile MR, Mullick AE, Saigusa T. Attenuation of accelerated renal cystogenesis in Pkd1 mice by renin-angiotensin system blockade. Am J Physiol Renal Physiol 2017; 314:F210-F218. [PMID: 29021226 DOI: 10.1152/ajprenal.00389.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The intrarenal renin angiotensin system (RAS) is activated in polycystic kidney disease. We have recently shown in the Pkd1 mouse that Gen 2 antisense oligonucleotide (ASO), which suppresses angiotensinogen (Agt) synthesis, is efficacious in slowing kidney cyst formation compared with lisinopril. The aim of this current study was to determine 1) if unilateral nephrectomy accelerates cystogenesis in Pkd1 mice (as previously shown in cilia knockout mice) and 2) whether Agt ASO can slow the progression in this accelerated cystic mouse model. Adult Pkd1 conditional floxed allele mice expressing cre were administered tamoxifen, resulting in global knockout of Pkd1. Three weeks after tamoxifen injection, mice underwent left unilateral nephrectomy. Mice were then treated with Agt ASO (75 mg/kg per week) or aliskiren (20 mg/kg per day)+Agt ASO or control for 8 wk. Unilateral nephrectomy accelerated kidney cyst formation compared with nonnephrectomized mice. Both Agt ASO and Aliskiren+Agt ASO treatments significantly reduced plasma and urinary Agt levels. Blood pressure was lowest in Aliskiren+Agt ASO mice among all treatment groups, and the control group had the highest blood pressure. All mice developed significant kidney cysts at 8 wk after nephrectomy, but Agt ASO and Aliskiren+Agt ASO groups had fewer kidney cysts than controls. Renal pAkt, pS6 levels, and apoptosis were significantly suppressed in those receiving Agt ASO compared with controls. These results indicate that suppressing Agt using an ASO slowed the progression of accelerated cystic kidney disease induced by unilateral nephrectomy in Pkd1 mice by suppressing intrarenal RAS, mammalian target of rapamycin pathway, and cell proliferation.
Collapse
Affiliation(s)
- Wayne R Fitzgibbon
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Yujing Dang
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Marlene A Bunni
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina
| | - Catalin F Baicu
- Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | - Michael R Zile
- Division of Cardiology, Medical University of South Carolina, Ralph H. Johnson Veterans Affairs Medical Center , Charleston, South Carolina
| | | | - Takamitsu Saigusa
- Divison of Nephrology, Medical University of South Carolina , Charleston, South Carolina.,Division of Nephrology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
29
|
Salih M, Bovée DM, Roksnoer LCW, Casteleijn NF, Bakker SJL, Gansevoort RT, Zietse R, Danser AHJ, Hoorn EJ. Urinary renin-angiotensin markers in polycystic kidney disease. Am J Physiol Renal Physiol 2017; 313:F874-F881. [PMID: 28747358 DOI: 10.1152/ajprenal.00209.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/05/2017] [Accepted: 07/20/2017] [Indexed: 11/22/2022] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD), activation of the renin-angiotensin aldosterone system (RAAS) may contribute to hypertension and disease progression. Although previous studies have focused on circulating RAAS components, preliminary evidence suggests that APDKD may increase urinary RAAS components. Therefore, our aim was to analyze circulating and urinary RAAS components in ADPKD. We cross-sectionally compared 60 patients with ADPKD with 57 patients with non-ADPKD chronic kidney disease (CKD). The two groups were matched by sex, estimated glomerular filtration rate (eGFR), blood pressure, and RAAS inhibitor use. Despite similar plasma levels of angiotensinogen and renin, urinary angiotensinogen and renin excretion were five- to sixfold higher in ADPKD (P < 0.001). These differences persisted when adjusting for group differences and were present regardless of RAAS inhibitor use. In multivariable analyses, ADPKD, albuminuria, and the respective plasma concentrations were independent predictors for urinary angiotensinogen and renin excretion. In ADPKD, both plasma and urinary renin correlated negatively with eGFR. Total kidney volume correlated with plasma renin and albuminuria but not with urinary renin or angiotensinogen excretions. Albuminuria correlated positively with urinary angiotensinogen and renin excretions in ADPKD and CKD. In three ADPKD patients who underwent nephrectomy, the concentrations of albumin and angiotensinogen were highest in plasma, followed by cyst fluid and urine; urinary renin concentrations were higher than cyst fluid. In conclusion, this study shows that, despite similar circulating RAAS component levels, higher urinary excretions of angiotensinogen and renin are a unique feature of ADPKD. Future studies should address the underlying mechanism and whether this may contribute to hypertension or disease progression in ADPKD.
Collapse
Affiliation(s)
- Mahdi Salih
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dominique M Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Lodi C W Roksnoer
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.,Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Niek F Casteleijn
- Department of Urology, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands; and
| | - Ronald T Gansevoort
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands; and
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands;
| |
Collapse
|
30
|
De Rechter S, Breysem L, Mekahli D. Is Autosomal Dominant Polycystic Kidney Disease Becoming a Pediatric Disorder? Front Pediatr 2017; 5:272. [PMID: 29326910 PMCID: PMC5742347 DOI: 10.3389/fped.2017.00272] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/04/2017] [Indexed: 12/15/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) affects 1 in 400 to 1,000 live births, making it the most common monogenic cause of renal failure. Although no definite cure is available yet, it is important to affect disease progression by influencing modifiable factors such as hypertension and proteinuria. Besides this symptomatic management, the only drug currently recommended in Europe for selected adult patients with rapid disease progression, is the vasopressin receptor antagonist tolvaptan. However, the question remains whether these preventive interventions should be initiated before extensive renal damage has occurred. As renal cyst formation and expansion begins early in life, frequently in utero, ADPKD should no longer be considered an adult-onset disease. Moreover, the presence of hypertension and proteinuria in affected children has been reported to correlate well with disease severity. Until now, it is controversial whether children at-risk for ADPKD should be tested for the presence of the disease, and if so, how this should be done. Herein, we review the spectrum of pediatric ADPKD and discuss the pro and contra of testing at-risk children and the challenges and unmet needs in pediatric ADPKD care.
Collapse
Affiliation(s)
- Stéphanie De Rechter
- PKD Lab, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Luc Breysem
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Djalila Mekahli
- PKD Lab, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Reddy BV, Chapman AB. The spectrum of autosomal dominant polycystic kidney disease in children and adolescents. Pediatr Nephrol 2017; 32:31-42. [PMID: 27034070 DOI: 10.1007/s00467-016-3364-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/02/2016] [Accepted: 03/02/2016] [Indexed: 12/19/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary renal disorder. It is characterized by the development of renal cysts and kidney enlargement and ultimately leads to renal failure typically in the sixth decade of life. Although most patients are asymptomatic until well into adulthood, renal cysts develop much earlier, often in utero. Significant renal anatomic and cystic expansion typically occurs before clinical manifestations in children and young adults with AKPKD. The cyst burden detected by imaging represents the minority of cyst burden, and renal and cardiovascular abnormalities are the most common manifestations in children with ADPKD. Here we review the molecular pathogenesis of ADPKD, discuss the screening, diagnosis and clinical manifestations of this renal disorder in childhood and adolescents and review treatment options and potential therapies currently being tested.
Collapse
Affiliation(s)
- Bharathi V Reddy
- University of Chicago, 5841, S. Maryland Avenue Suite S-511, MC 5100, Chicago, IL, 60637, USA.
| | - Arlene B Chapman
- University of Chicago, 5841, S. Maryland Avenue Suite S-511, MC 5100, Chicago, IL, 60637, USA
| |
Collapse
|
32
|
Haller H, Park JK, Lindschau C, Meyer M, Menne J. Intrarenal renin-angiotensin system — important player of the local milieu. J Renin Angiotensin Aldosterone Syst 2016; 7:122-5. [PMID: 17083066 DOI: 10.3317/jraas.2006.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Affiliation(s)
- Hermann Haller
- Department of Nephrology, Hannover Medical School, Germany
| | | | | | | | | |
Collapse
|
33
|
Lai S, Petramala L, Mastroluca D, Petraglia E, Di Gaeta A, Indino E, Panebianco V, Ciccariello M, Shahabadi HH, Galani A, Letizia C, D’Angelo AR. Hyperaldosteronism and cardiovascular risk in patients with autosomal dominant polycystic kidney disease. Medicine (Baltimore) 2016; 95:e4175. [PMID: 27442639 PMCID: PMC5265756 DOI: 10.1097/md.0000000000004175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/04/2016] [Accepted: 06/14/2016] [Indexed: 01/22/2023] Open
Abstract
Hypertension is commonly associated with autosomal dominant polycystic kidney disease (ADPKD), often discovered before the onset of renal failure, albeit the pathogenetic mechanisms are not well elucidated. Hyperaldosteronism in ADPKD may contribute to the development of insulin resistance and endothelial dysfunction, and progression of cardiorenal disease. The aim of study was to evaluate the prevalence of primary aldosteronism (PA) in ADPKD patients and identify some surrogate biomarkers of cardiovascular risk.We have enrolled 27 hypertensive ADPKD patients with estimated glomerular filtration rate (eGFR) ≥ 60 mL/min, evaluating the renin-angiotensin-aldosterone system (RAAS), inflammatory indexes, nutritional status, homocysteine (Hcy), homeostasis model assessment-insulin resistance (HOMA-IR), mineral metabolism, microalbuminuria, and surrogate markers of atherosclerosis [carotid intima media thickness (cIMT), ankle/brachial index (ABI), flow mediated dilation (FMD), renal resistive index (RRI) and left ventricular mass index (LVMI)]. Furthermore, we have carried out the morpho-functional magnetic resonance imaging (MRI) with high-field 3 T Magnetom Avanto.We have divided patients into group A, with normal plasma aldosterone concentration (PAC) and group B with PA, present in 9 (33%) of overall ADPKD patients. Respect to group A, group B showed a significant higher mean value of LVMI, HOMA-IR and Hcy (P = 0.001, P = 0.004, P = 0.018; respectively), and a lower value of FMD and 25-hydroxyvitamin D (25-OH-VitD) (P = 0.037, P = 0.019; respectively) with a higher prevalence of non-dipper pattern at Ambulatory Blood Pressure Monitoring (ABPM) (65% vs 40%, P < 0.05) at an early stage of the disease.In this study, we showed a high prevalence of PA in ADPKD patients, associated to higher LVMI, HOMA-IR, Hcy, lower FMD, and 25-OH-VitD, considered as surrogate markers of atherosclerosis, compared to ADPKD patients with normal PAC values. Our results indicate a higher overall cardiovascular risk in ADPKD patients with inappropriate aldosterone secretion, and a screening for PA in all patients with ADPKD is recommended.
Collapse
Affiliation(s)
- Silvia Lai
- Department of Clinical Medicine, Sapienza University of Rome, Rome
| | - Luigi Petramala
- Department of Internal Medicine and Medical Specialities, Sapienza University of Rome, Rome
| | - Daniela Mastroluca
- Department of Internal Medicine and Medical Specialities, Sapienza University of Rome, Rome
| | | | - Alessandro Di Gaeta
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome
| | - Elena Indino
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome
| | - Valeria Panebianco
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome
| | - Mauro Ciccariello
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome
| | - Hossein H. Shahabadi
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome
| | - Alessandro Galani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Claudio Letizia
- Department of Internal Medicine and Medical Specialities, Sapienza University of Rome, Rome
| | - Anna Rita D’Angelo
- Department of Obstetrical-Gynecological Sciences and Urologic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
34
|
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a signalopathy of renal tubular epithelial cells caused by naturally occurring mutations in two distinct genes, polycystic kidney disease 1 (PKD1) and 2 (PKD2). Genetic variants in PKD1, which encodes the polycystin-1 (PC-1) protein, remain the predominant factor associated with the pathogenesis of nearly two-thirds of all patients diagnosed with PKD. Although the relationship between defective PC-1 with renal cystic disease initiation and progression remains to be fully elucidated, there are numerous clinical studies that have focused upon the control of effector systems involving heterotrimeric G protein regulation. A major regulator in the activation state of heterotrimeric G proteins are G protein-coupled receptors (GPCRs), which are defined by their seven transmembrane-spanning regions. PC-1 has been considered to function as an unconventional GPCR, but the mechanisms by which PC-1 controls signal processing, magnitude, or trafficking through heterotrimeric G proteins remains to be fully known. The diversity of heterotrimeric G protein signaling in PKD is further complicated by the presence of non-GPCR proteins in the membrane or cytoplasm that also modulate the functional state of heterotrimeric G proteins within the cell. Moreover, PC-1 abnormalities promote changes in hormonal systems that ultimately interact with distinct GPCRs in the kidney to potentially amplify or antagonize signaling output from PC-1. This review will focus upon the canonical and noncanonical signaling pathways that have been described in PKD with specific emphasis on which heterotrimeric G proteins are involved in the pathological reorganization of the tubular epithelial cell architecture to exacerbate renal cystogenic pathways.
Collapse
Affiliation(s)
- Taketsugu Hama
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
35
|
Saigusa T, Bell PD. Molecular pathways and therapies in autosomal-dominant polycystic kidney disease. Physiology (Bethesda) 2016; 30:195-207. [PMID: 25933820 DOI: 10.1152/physiol.00032.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is the most prevalent inherited renal disease, characterized by multiple cysts that can eventually lead to kidney failure. Studies investigating the role of primary cilia and polycystins have significantly advanced our understanding of the pathogenesis of PKD. This review will present clinical and basic aspects of ADPKD, review current concepts of PKD pathogenesis, evaluate potential therapeutic targets, and highlight challenges for future clinical studies.
Collapse
Affiliation(s)
- Takamitsu Saigusa
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina; and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - P Darwin Bell
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, South Carolina; and Ralph Johnson VA Medical Center, Charleston, South Carolina
| |
Collapse
|
36
|
Ramanathan G, Elumalai R, Periyasamy S, Lakkakula BVKS. Renin gene rs1464816 polymorphism contributes to chronic kidney disease progression in ADPKD. J Biomed Sci 2016; 23:1. [PMID: 26753721 PMCID: PMC4710007 DOI: 10.1186/s12929-015-0217-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 12/28/2015] [Indexed: 01/02/2023] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic disorder and is a common genetic cause of chronic renal failure in children and adults. The enzyme renin plays a key role in the RAAS cascade and an important role in the development of hypertension and progression of renal disease in ADPKD. The present study is aimed to investigate the potential modifier effect of REN gene polymorphisms on the progression of chronic kidney disease (CKD) in ADPKD. Methods We analyzed 102 ADPKD patients and 106 healthy controls from the same geographic area. FRET-based KASPar single-nucleotide polymorphism (SNP) genotyping assays for REN gene tag-SNPs (rs2887284, rs2368564, rs1464816, rs7521667, rs10900555, rs6693954, rs6676670 and rs11571078) were performed. Cochran-Armitage trend test was used to assess the potential associations between these polymorphisms and CKD stages. Haplotype frequencies and LD measures were estimated by using the software Haploview. Mantel-Haenszel stratified analysis was used to explore confounding and interaction effects of these polymorphisms. Results Of the eight tag-SNPs genotyped, the rs10900555 polymorphism deviated from the Hardy-Weinberg equilibrium in controls. The presence of ADPKD in general was not significantly associated with the REN tag-SNPs included in this study. Linkage disequilibrium analysis yielded three haplotype blocks and the haplotypes of the respective blocks are not statistically different between ADPKD and controls. In multivariate analysis, the rs1464816 TG genotype showed a significant association with the advancement of CKD in ADPKD (OR = 4.80; 95 % CI = 1.30–17.82; p = 0.019). Conclusions The present study provides evidence that the rs1464816 polymorphism in REN is associated with CKD progression in ADPKD. Electronic supplementary material The online version of this article (doi:10.1186/s12929-015-0217-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | - Bhaskar V K S Lakkakula
- Department of Biomedical Sciences, Sri Ramachandra University, Chennai, India. .,Genetic Lab, Department of Biochemistry, Sickle Cell Institute Chhattisgarh, Pt. JNM Medical College, Raipur, 492001, CG, India.
| |
Collapse
|
37
|
Eriguchi M, Yotsueda R, Torisu K, Kawai Y, Hasegawa S, Tanaka S, Noguchi H, Masutani K, Kitazono T, Tsuruya K. Assessment of urinary angiotensinogen as a marker of podocyte injury in proteinuric nephropathies. Am J Physiol Renal Physiol 2015; 310:F322-33. [PMID: 26632605 DOI: 10.1152/ajprenal.00260.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/01/2015] [Indexed: 01/13/2023] Open
Abstract
Urinary protein (UP) is widely used as a clinical marker for podocyte injury; however, not all proteinuric nephropathies fit this model. We previously described the elevation of urinary angiotensinogen (AGT) accompanied by AGT expression by injured podocytes in a nitric oxide inhibition rat model (Eriguchi M, Tsuruya K, Haruyama N, Yamada S, Tanaka S, Suehiro T, Noguchi H, Masutani K, Torisu K, Kitazono T. Kidney Int 87: 116-127, 2015). In this report, we performed the human and animal studies to examine the significance and origin of urinary AGT. In the human study, focal segmental glomerulosclerosis (FSGS) patients presented with higher levels of urinary AGT, corrected by UP, than minimal-change disease (MCD) patients. Furthermore, AGT was evident in podocin-negative glomerular segmental lesions. We also tested two different nephrotic models induced by puromycin aminonucleoside in Wistar rats. The urinary AGT/UP ratio and AGT protein and mRNA expression in sieved glomeruli from FSGS rats were significantly higher than in MCD rats. The presence of AGT at injured podocytes in FSGS rats was detected by immunohistochemistry and immunoelectron microscopy. Finally, we observed the renal tissue and urinary metabolism of exogenous injected human recombinant AGT (which is not cleaved by rodent renin) in FSGS and control rats. Significant amounts of human AGT were detected in the urine of FSGS rats, but not of control rats. Immunostaining for rat and human AGT identified that only rat AGT was detected in injured podocytes, and filtered human AGT was seen in superficial proximal tubules, but not in injured podocytes, suggesting AGT generation by injured podocytes. In conclusion, the urinary AGT/UP ratio represents a novel specific marker of podocyte injury.
Collapse
Affiliation(s)
- Masahiro Eriguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Ryusuke Yotsueda
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Kumiko Torisu
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Yasuhiro Kawai
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Shoko Hasegawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Shigeru Tanaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Hideko Noguchi
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Kosuke Masutani
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and
| | - Kazuhiko Tsuruya
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; and Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
38
|
Olowu WA. Pre-treatment considerations in childhood hypertension due to chronic kidney disease. World J Nephrol 2015; 4:500-510. [PMID: 26558187 PMCID: PMC4635370 DOI: 10.5527/wjn.v4.i5.500] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/25/2015] [Accepted: 10/08/2015] [Indexed: 02/06/2023] Open
Abstract
Hypertension (HTN) develops very early in childhood chronic kidney disease (CKD). It is linked with rapid progression of kidney disease, increased morbidity and mortality hence the imperative to start anti-hypertensive medication when blood pressure (BP) is persistently > 90th percentile for age, gender, and height in non-dialyzing hypertensive children with CKD. HTN pathomechanism in CKD is multifactorial and complexly interwoven. The patient with CKD-associated HTN needs to be carefully evaluated for co-morbidities that frequently alter the course of the disease as successful treatment of HTN in CKD goes beyond life style modification and anti-hypertensive therapy alone. Chronic anaemia, volume overload, endothelial dysfunction, arterial media calcification, and metabolic derangements like secondary hyperparathyroidism, hyperphosphataemia, and calcitriol deficiency are a few co-morbidities that may cause or worsen HTN in CKD. It is important to know if the HTN is caused or made worse by the toxic effects of medications like erythropoietin, cyclosporine, tacrolimus, corticosteroids and non-steroidal anti-inflammatory drugs. Poor treatment response may be due to any of these co-morbidities and medications. A satisfactory hypertensive CKD outcome, therefore, depends very much on identifying and managing these co-morbid conditions and HTN promoting medications promptly and appropriately. This review attempts to point attention to factors that may affect successful treatment of the hypertensive CKD child and how to attain the desired therapeutic BP target.
Collapse
|
39
|
Saigusa T, Dang Y, Mullick AE, Yeh ST, Zile MR, Baicu CF, Bell PD. Suppressing angiotensinogen synthesis attenuates kidney cyst formation in a Pkd1 mouse model. FASEB J 2015; 30:370-9. [PMID: 26391272 DOI: 10.1096/fj.15-279299] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/08/2015] [Indexed: 01/13/2023]
Abstract
Activation of the intrarenal renin angiotensin system (RAS) is believed to play an important role in the development of hypertension and cystogenesis in autosomal dominant polycystic kidney disease (ADPKD). Results of clinical studies testing RAS inhibitors in slowing the progression of cystic disease in ADPKD are inconclusive, and we hypothesized that current RAS inhibitors do not adequately suppress intrarenal RAS. For this study, we compared a novel Gen 2 antisense oligonucleotide (ASO) that inhibits angiotensinogen (Agt) synthesis to lisinopril in adult conditional Pkd1 systemic-knockout mice, a model of ADPKD. Six weeks after Pkd1 global gene knockout, the mice were treated with Agt-ASO (66 mg/kg/wk), lisinopril (100 mg/kg/d), PBS (control), or scrambled ASO (66 mg/kg/wk) for 10 wk, followed by tissue collection. Agt ASO resulted in significant reduction in plasma, liver, and kidney Agt, and increased kidney renin compared with control treatments. Kidneys from Agt-ASO-treated mice were not as enlarged and showed reduced cystic volume compared with lisinopril or control treatments. Blood pressure was better controlled with lisinopril than with Agt-ASO. Agt-ASO suppressed cell proliferation in both cystic and noncystic cells compared with lisinopril and control treatments. However, Agt-ASO did not reduce cell proliferation in liver, which indicates that Agt-ASO targets cell signaling pathways that specifically suppresses cystogenesis in the kidney. These data suggest that Agt-ASO effectively attenuates intrarenal RAS and therefore can be a novel and effective agent for treating ADPKD.
Collapse
Affiliation(s)
- Takamitsu Saigusa
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - Yujing Dang
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - Adam E Mullick
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - Steve T Yeh
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - Michael R Zile
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - Catalin F Baicu
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| | - P Darwin Bell
- *Division of Nephrology and Division of Cardiology, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA; and Isis Pharmaceuticals, Carlsbad, California, USA
| |
Collapse
|
40
|
Increased urinary Angiotensinogen/Creatinine (AGT/Cr) ratio may be associated with reduced renal function in autosomal dominant polycystic kidney disease patients. BMC Nephrol 2015; 16:86. [PMID: 26092580 PMCID: PMC4475321 DOI: 10.1186/s12882-015-0075-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 05/21/2015] [Indexed: 01/31/2023] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common hereditary kidney diseases that frequently result in renal failure. In this cross-sectional observational cohort study, we evaluated urinary angiotensinogen (AGT) as a potential biomarker to assess renal function in ADPKD. Methods Urinary AGT was measured in 233 ADPKD patients and its association with estimated glomerular filtration rate (eGFR) and height-adjusted total kidney volume (htTKV) were evaluated. The localization of AGT and other renin-angiotensin system (RAS)-related molecules were identified using immunohistochemistry in human ADPKD tissues. Results Baseline urinary AGT/Cr was negatively correlated with CKD-EPI eGFR (r2
= 0.162, P < 0.001) and positively correlated with htTKV (r2 = 0.107, P < 0.001). Both urinary AGT/Cr and plasma renin activity levels were significantly elevated in hypertensive ADPKD patients. Among hypertensive subjects, urinary AGT/Cr was significantly increased in the advanced CKD stages (III-V) compared to early CKD stages (I-II) (28.6 ± 60.3 vs. 93.2 ± 139.3 μg/g, P < 0.001). Immunohistochemical study showed strong expression of AGT along the cyst-lining epithelial cells as well as the nearby compressed tubular epithelial cells. Conclusions Our results suggested that urinary AGT/Cr may be a valuable biomarker for renal damage in ADPKD since intrarenal ischemic insults induced by cyst growth and subsequent intrarenal RAS activation may play a potential role in the development of hypertension and renal dysfunction in ADPKD. Electronic supplementary material The online version of this article (doi:10.1186/s12882-015-0075-8) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Saigusa T, Dang Y, Bunni MA, Amria MY, Steele SL, Fitzgibbon WR, Bell PD. Activation of the intrarenal renin-angiotensin-system in murine polycystic kidney disease. Physiol Rep 2015; 3:3/5/e12405. [PMID: 25999403 PMCID: PMC4463833 DOI: 10.14814/phy2.12405] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The mechanism for early hypertension in polycystic kidney disease (PKD) has not been elucidated. One potential pathway that may contribute to the elevation in blood pressure in PKD is the activation of the intrarenal renin-angiotensin-system (RAS). For example, it has been shown that kidney cyst and cystic fluid contains renin, angiotensin II (AngII), and angiotensinogen (Agt). Numerous studies suggest that ciliary dysfunction plays an important role in PKD pathogenesis. However, it is unknown whether the primary cilium affects the intrarenal RAS in PKD. The purpose of this study was to determine whether loss of cilia or polycystin 1 (PC1) increases intrarenal RAS in mouse model of PKD. Adult Ift88 and Pkd1 conditional floxed allele mice with or without cre were administered tamoxifen to induce global knockout of the gene. Three months after tamoxifen injection, kidney tissues were examined by histology, immunofluorescence, western blot, and mRNA to assess intrarenal RAS components. SV40 immortalized collecting duct cell lines from hypomorphic Ift88 mouse were used to assess intrarenal RAS components in collecting duct cells. Mice without cilia and PC1 demonstrated increased kidney cyst formation, systolic blood pressure, prorenin, and kidney and urinary angiotensinogen levels. Interestingly immunofluorescence study of the kidney revealed that the prorenin receptor was localized to the basolateral membrane of principal cells in cilia (−) but not in cilia (+) kidneys. Collecting duct cAMP responses to AngII administration was greater in cilia (−) vs. cilia (+) cells indicating enhanced intrarenal RAS activity in the absence of cilia. These data suggest that in the absence of cilia or PC1, there is an upregulation of intrarenal RAS components and activity, which may contribute to elevated blood pressure in PKD.
Collapse
Affiliation(s)
- Takamitsu Saigusa
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - Yujing Dang
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - Marlene A Bunni
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - May Y Amria
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - Stacy L Steele
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - Wayne R Fitzgibbon
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, Medical University of South Carolina Charleston SC and Ralph Johnson VA Medical Center, Charleston, South Carolina
| |
Collapse
|
42
|
Ravichandran K, Ozkok A, Wang Q, Mullick AE, Edelstein CL. Antisense-mediated angiotensinogen inhibition slows polycystic kidney disease in mice with a targeted mutation in Pkd2. Am J Physiol Renal Physiol 2014; 308:F349-57. [PMID: 25537744 DOI: 10.1152/ajprenal.00478.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Renal cyst enlargement is associated with the activation of both the circulating and intrarenal renin-angiotensin systems. Angiotensinogen (AGT) is the substrate for renin. The aim of the present study was to determine the effect of AGT inhibition on renal cyst enlargement. An AGT antisense oligonucleotide (ASO) that selectively inhibits AGT mRNA was injected once weekly in PKD2WS25 mice [an orthologous model of human autosmal dominant polycystic kidney disease (PKD) involving mutation of the Pkd2 gene] from 4 to 16 wk of age. The AGT ASO resulted in a 40% decrease in AGT RNA in the kidney, a 60% decrease in AGT RNA in the liver, and a significant decrease in AGT protein in the kidney and serum. The AGT ASO resulted in a significant decrease in kidney size, cyst volume density, and blood urea nitrogen. The AGT ASO resulted in a significant decrease in transforming growth factor-β and interstitial fibrosis in the kidney. Mice treated with the AGT ASO had a significant decrease in proinflammatory cytokines [chemokine (C-X-C motif) ligand (CXCL)1 and IL-12] in the kidney. Cluster of differentiation (CD)36 is a scavenger receptor found on tubular cells that can activate the renin-angiotensin system. Administration of a CD36 ASO had no effect on PKD and kidney function, suggesting that the effect of the AGT ASO is independent of CD36. In summary, AGT inhibition resulted in significant decreases in kidney size and cyst volume and an improvement in kidney function in PKD mice. The AGT ASO resulted in a decrease in transforming growth factor-β, interstitial fibrosis, and the proinflammatory cytokines CXCL1 and IL-12 in the kidney.
Collapse
Affiliation(s)
- Kameswaran Ravichandran
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, Colorado; and
| | - Abdullah Ozkok
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, Colorado; and
| | - Qian Wang
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, Colorado; and
| | | | - Charles L Edelstein
- Division of Renal Diseases and Hypertension, University of Colorado at Denver, Aurora, Colorado; and
| |
Collapse
|
43
|
Kocyigit I, Eroglu E. Urinary angiotensinogen can be useful for screening patients with polycystic kidney disease. Kidney Int 2014; 85:714. [PMID: 24583996 DOI: 10.1038/ki.2013.515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Ismail Kocyigit
- Department of Nephrology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Eray Eroglu
- Department of Internal Medicine, Erciyes University Medical Faculty, Kayseri, Turkey
| |
Collapse
|
44
|
Torres VE, Abebe KZ, Chapman AB, Schrier RW, Braun WE, Steinman TI, Winklhofer FT, Brosnahan G, Czarnecki PG, Hogan MC, Miskulin DC, Rahbari-Oskoui FF, Grantham JJ, Harris PC, Flessner MF, Moore CG, Perrone RD. Angiotensin blockade in late autosomal dominant polycystic kidney disease. N Engl J Med 2014; 371:2267-76. [PMID: 25399731 PMCID: PMC4284824 DOI: 10.1056/nejmoa1402686] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypertension develops early in patients with autosomal dominant polycystic kidney disease (ADPKD) and is associated with disease progression. The renin-angiotensin-aldosterone system (RAAS) is implicated in the pathogenesis of hypertension in patients with ADPKD. Dual blockade of the RAAS may circumvent compensatory mechanisms that limit the efficacy of monotherapy with an angiotensin-converting-enzyme (ACE) inhibitor or angiotensin II-receptor blocker (ARB). METHODS In this double-blind, placebo-controlled trial, we randomly assigned 486 patients, 18 to 64 years of age, with ADPKD (estimated glomerular filtration rate [GFR], 25 to 60 ml per minute per 1.73 m(2) of body-surface area) to receive an ACE inhibitor (lisinopril) and placebo or lisinopril and an ARB (telmisartan), with the doses adjusted to achieve a blood pressure of 110/70 to 130/80 mm Hg. The composite primary outcome was the time to death, end-stage renal disease, or a 50% reduction from the baseline estimated GFR. Secondary outcomes included the rates of change in urinary aldosterone and albumin excretion, frequency of hospitalizations for any cause and for cardiovascular causes, incidence of pain, frequency of ADPKD-related symptoms, quality of life, and adverse study-medication effects. Patients were followed for 5 to 8 years. RESULTS There was no significant difference between the study groups in the incidence of the composite primary outcome (hazard ratio with lisinopril-telmisartan, 1.08; 95% confidence interval, 0.82 to 1.42). The two treatments controlled blood pressure and lowered urinary aldosterone excretion similarly. The rates of decline in the estimated GFR, urinary albumin excretion, and other secondary outcomes and adverse events, including hyperkalemia and acute kidney injury, were also similar in the two groups. CONCLUSIONS Monotherapy with an ACE inhibitor was associated with blood-pressure control in most patients with ADPKD and stage 3 chronic kidney disease. The addition of an ARB did not alter the decline in the estimated GFR. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; HALT-PKD [Study B] ClinicalTrials.gov number, NCT01885559.).
Collapse
Affiliation(s)
- Vicente E Torres
- From the Mayo Clinic College of Medicine, Rochester, MN (V.E.T., M.C.H., P.C.H.); University of Pittsburgh School of Medicine, Pittsburgh (K.Z.A., C.G.M.); Emory University School of Medicine, Atlanta (A.B.C., F.F.R.-O.); University of Colorado Health Sciences Center, Denver (R.W.S., G.B.); Cleveland Clinic, Cleveland (W.E.B.); Beth Israel Deaconess Medical Center (T.I.S., P.G.C.) and Tufts Medical Center (D.C.M., R.D.P.) - both in Boston; Kansas University Medical Center, Kansas City (F.T.W., J.J.G.) and the National Institutes of Health, Bethesda, MD (M.F.F.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ow CPC, Abdelkader A, Hilliard LM, Phillips JK, Evans RG. Determinants of renal tissue hypoxia in a rat model of polycystic kidney disease. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1207-15. [DOI: 10.1152/ajpregu.00202.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Renal tissue oxygen tension (Po2) and its determinants have not been quantified in polycystic kidney disease (PKD). Therefore, we measured kidney tissue Po2 in the Lewis rat model of PKD (LPK) and in Lewis control rats. We also determined the relative contributions of altered renal oxygen delivery and consumption to renal tissue hypoxia in LPK rats. Po2 of the superficial cortex of 11- to 13-wk-old LPK rats, measured by Clark electrode with the rat under anesthesia, was higher within the cysts (32.8 ± 4.0 mmHg) than the superficial cortical parenchyma (18.3 ± 3.5 mmHg). Po2 in the superficial cortical parenchyma of Lewis rats was 2.5-fold greater (46.0 ± 3.1 mmHg) than in LPK rats. At each depth below the cortical surface, tissue Po2 in LPK rats was approximately half that in Lewis rats. Renal blood flow was 60% less in LPK than in Lewis rats, and arterial hemoglobin concentration was 57% less, so renal oxygen delivery was 78% less. Renal venous Po2 was 38% less in LPK than Lewis rats. Sodium reabsorption was 98% less in LPK than Lewis rats, but renal oxygen consumption did not significantly differ between the two groups. Thus, in this model of PKD, kidney tissue is severely hypoxic, at least partly because of deficient renal oxygen delivery. Nevertheless, the observation of similar renal oxygen consumption, despite markedly less sodium reabsorption, in the kidneys of LPK compared with Lewis rats, indicates the presence of inappropriately high oxygen consumption in the polycystic kidney.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Department of Physiology Monash University, Melbourne, Australia; and
| | - Amany Abdelkader
- Department of Physiology Monash University, Melbourne, Australia; and
| | | | | | - Roger G. Evans
- Department of Physiology Monash University, Melbourne, Australia; and
| |
Collapse
|
46
|
Fonseca JM, Bastos AP, Amaral AG, Sousa MF, Souza LE, Malheiros DM, Piontek K, Irigoyen MC, Watnick TJ, Onuchic LF. Renal cyst growth is the main determinant for hypertension and concentrating deficit in Pkd1-deficient mice. Kidney Int 2014; 85:1137-50. [PMID: 24429399 PMCID: PMC4510986 DOI: 10.1038/ki.2013.501] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/27/2013] [Accepted: 10/03/2013] [Indexed: 01/13/2023]
Abstract
We have bred a Pkd1 floxed allele with a nestin-Cre expressing line to generate cystic mice with preserved glomerular filtration rate to address the pathogenesis of complex autosomal dominant polycystic kidney disease (ADPKD) phenotypes. Hypertension affects about 60% of these patients before loss of renal function, leading to significant morbimortality. Cystic mice were hypertensive at 5 and 13 weeks of age, a phenotype not seen in noncystic controls and Pkd1-haploinsufficient animals that do not develop renal cysts. Fractional sodium excretion was reduced in cystic mice at these ages. Angiotensinogen gene expression was higher in cystic than noncystic kidneys at 18 weeks, while ACE and the AT1 receptor were expressed in renal cyst epithelia. Cystic animals displayed increased renal cAMP, cell proliferation, and apoptosis. At 24 weeks, mean arterial pressure and fractional sodium excretion did not significantly differ between the cystic and noncystic groups, whereas cardiac mass increased in cystic mice. Renal concentrating deficit is also an early finding in ADPKD. Maximum urine osmolality and urine nitrite excretion were reduced in 10-13- and 24-week-old cystic mice, deficits not found in haploinsufficient and noncystic controls. A trend of higher plasma vasopressin was observed in cystic mice. Thus, cyst growth most probably plays a central role in early-stage ADPKD-associated hypertension, with activation of the intrarenal renin-angiotensin system as a key mechanism. Cyst expansion is also likely essential for the development of the concentrating deficit in this disease. Our findings are consistent with areas of reduced perfusion in the kidneys of patients with ADPKD.
Collapse
Affiliation(s)
- Jonathan M Fonseca
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Ana P Bastos
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Andressa G Amaral
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Mauri F Sousa
- Department of Medicine, Federal University of Goiás School of Medicine, Goiânia, Brazil
| | - Leandro E Souza
- Department of Cardiopneumology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Denise M Malheiros
- Department of Pathology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Klaus Piontek
- Department of Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Maria C Irigoyen
- Department of Cardiopneumology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Terry J Watnick
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luiz F Onuchic
- 1] Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil [2] Center for Cellular and Molecular Studies and Therapy (NETCEM), University of São Paulo, São Paulo, Brazil
| |
Collapse
|
47
|
Torra R. Tratamiento de la poliquistosis renal autosómica dominante. Med Clin (Barc) 2014; 142:73-9. [DOI: 10.1016/j.medcli.2013.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 09/04/2013] [Accepted: 09/12/2013] [Indexed: 01/22/2023]
|
48
|
From bone abnormalities to mineral metabolism dysregulation in autosomal dominant polycystic kidney disease. Pediatr Nephrol 2013; 28:2089-96. [PMID: 23340856 DOI: 10.1007/s00467-012-2384-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 11/27/2012] [Accepted: 11/28/2012] [Indexed: 01/02/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of kidney failure. It is a systemic disorder, not only affecting the kidneys, but also associated with cyst formation in other organs such as the liver, spleen, pancreas, and seminal vesicles. Other extra-renal symptoms may consist of intracranial arterial aneurysms, cardiac valvular defects, abdominal and inguinal hernias and colonic diverticulosis. Very little is known regarding bone involvement in ADPKD; however, recent evidence has revealed the potential role of fibroblast growth factor 23 (FGF23). FGF23 is an endocrine fibroblast growth factor acting in the kidney as a phosphaturic hormone and a suppressor of active vitamin D with key effects on the bone/kidney/parathyroid axis, and has been shown to increase in patients with ADPKD, even with normal renal function. The aim of this review is to provide an overview of bone and mineral abnormalities found in experimental models and in patients with ADPKD, and to discuss the possible role of FGF23 in this disease.
Collapse
|
49
|
Changing referral characteristics of patients with autosomal dominant polycystic kidney disease. Am J Med 2013; 126:832.e7-832.e11. [PMID: 23830538 DOI: 10.1016/j.amjmed.2012.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 12/10/2012] [Accepted: 12/10/2012] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Autosomal dominant polycystic kidney disease is the most frequent life-threatening hereditary disease. The study objective was to assess whether the clinical characteristics of patients with autosomal dominant polycystic kidney disease who are referred to a major autosomal dominant polycystic kidney disease center have changed over time. METHODS The clinical characteristics of patients with autosomal dominant polycystic kidney disease were compared between period A (1961-1990) and period B (1991-2011). The study took place at the Autosomal Dominant Polycystic Kidney Disease Center at the University of Colorado. A total of 837 patients referred with autosomal dominant polycystic kidney disease were included. Blood pressure control and renin-angiotensin-aldosterone system inhibition were instituted. Renal function, blood pressure, end-stage renal disease, and mortality were analyzed. RESULTS The results in period B compared with period A demonstrated an earlier age of autosomal dominant polycystic kidney disease diagnosis (29 vs 35 years, P < .001), lower mean blood pressure (129/82 vs 142/91 mm Hg, P < .001), better estimated glomerular filtration rate (63.6 vs 44.6 mL/min, P < .001), and more therapy with angiotensin-converting enzyme inhibition (42.5% vs 13.6%, P < .001). Time from birth to end-stage renal disease (52.8 ± 0.6 vs 49.1 ± 0.6 years, P < .001) and birth to death (63.5 ± 1.5 years vs 57.2 ± 1.0 years, P < .001) was longer in period B compared with period A when adjusted for age at diagnosis, sex, and estimated glomerular filtration rate. The study was retrospective, which is a limitation. CONCLUSIONS In period B, there was significantly better blood pressure control, more renin-angiotensin-aldosterone system inhibition, better preservation of renal function, and a longer period from birth to end-stage renal disease and death.
Collapse
|
50
|
Huang JL, Woolf AS, Long DA. Angiogenesis and autosomal dominant polycystic kidney disease. Pediatr Nephrol 2013; 28:1749-55. [PMID: 22990303 DOI: 10.1007/s00467-012-2305-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 08/20/2012] [Accepted: 08/21/2012] [Indexed: 10/27/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of multiple cysts that in many cases result in end-stage renal disease. Current strategies to reduce cyst progression in ADPKD focus on modulating cell turnover, fluid secretion, and vasopressin signalling; but an alternative approach may be to target pathways providing "general support" for cyst growth, such as surrounding blood vessels. This could be achieved by altering the expression of growth factors involved in vascular network formation, such as the vascular endothelial growth factor (VEGF) and angiopoietin families. We highlight the evidence that blood vessels and vascular growth factors play a role in ADPKD progression. Recent experiments manipulating VEGF in ADPKD are described, and we discuss how alternative strategies to manipulate angiogenesis may be used in the future as a novel treatment for ADPKD.
Collapse
Affiliation(s)
- Jennifer L Huang
- Nephro-Urology Unit, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | | | | |
Collapse
|