1
|
Sinha SK, Nicholas SB. Pathomechanisms of Diabetic Kidney Disease. J Clin Med 2023; 12:7349. [PMID: 38068400 PMCID: PMC10707303 DOI: 10.3390/jcm12237349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 03/15/2024] Open
Abstract
The worldwide occurrence of diabetic kidney disease (DKD) is swiftly rising, primarily attributed to the growing population of individuals affected by type 2 diabetes. This surge has been transformed into a substantial global concern, placing additional strain on healthcare systems already grappling with significant demands. The pathogenesis of DKD is intricate, originating with hyperglycemia, which triggers various mechanisms and pathways: metabolic, hemodynamic, inflammatory, and fibrotic which ultimately lead to renal damage. Within each pathway, several mediators contribute to the development of renal structural and functional changes. Some of these mediators, such as inflammatory cytokines, reactive oxygen species, and transforming growth factor β are shared among the different pathways, leading to significant overlap and interaction between them. While current treatment options for DKD have shown advancement over previous strategies, their effectiveness remains somewhat constrained as patients still experience residual risk of disease progression. Therefore, a comprehensive grasp of the molecular mechanisms underlying the onset and progression of DKD is imperative for the continued creation of novel and groundbreaking therapies for this condition. In this review, we discuss the current achievements in fundamental research, with a particular emphasis on individual factors and recent developments in DKD treatment.
Collapse
Affiliation(s)
- Satyesh K. Sinha
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
- College of Medicine, Charles R Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| |
Collapse
|
2
|
Jain A, Jain I. ET-traps: Potential therapeutics for preeclampsia. Drug Discov Today 2023; 28:103787. [PMID: 37742912 DOI: 10.1016/j.drudis.2023.103787] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
Elevated endothelin-1 (ET-1) has been implicated in several diseases including preeclampsia, where it causes the induction of hypertension, oxidative stress, endoplasmic reticulum stress, microvascular dysfunction and tissue damage in different organs. ET-traps are Fc-fusion proteins with a design based on the physiological receptors of ET-1. This paper discusses the potential use of ET-traps as a therapeutic for preeclampsia. ET-traps potently bind and sequester pathologically elevated ET-1 to significantly reduce different markers of pathology to non-disease levels with no toxicity.
Collapse
Affiliation(s)
- Arjun Jain
- ET-traps, Cambridge, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, UK; Accelerate Cambridge, Judge Business School, University of Cambridge, UK; Imperial College London, UK
| | - Ira Jain
- ET-traps, Cambridge, UK; Indian Institute of Management, Ahmedabad, India; National University of Singapore, Singapore.
| |
Collapse
|
3
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Arai H, Yamamoto S, Matsubara T, Miyake T, Tochio A, Mii A, Shimizu A, Minamiguchi S, Muso E, Yanagita M. Focal Segmental Sclerosis Associated with the Novel Multi-tyrosine Kinase Inhibitor Ponatinib. Intern Med 2023; 62:2693-2698. [PMID: 36642524 PMCID: PMC10569927 DOI: 10.2169/internalmedicine.1283-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023] Open
Abstract
Ponatinib is a novel multi-tyrosine kinase inhibitor (TKI) with potent inhibitory activity against refractory chronic myeloid leukemia (CML). Despite its high clinical efficacy, ponatinib induces various adverse events due to its multi-target characteristic. However, renal complications associated with ponatinib are rare. A 76-year-old woman had a history of chronic myeloid leukemia (CML) resistant to imatinib and nilotinib. Our patient developed proteinuria and renal function deterioration during treatment with ponatinib but not with imatinib or nilotinib. We herein report the first case of a patient with secondary focal segmental glomerulosclerosis (FSGS) with partial glomerular collapse induced by ponatinib treatment.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Shinya Yamamoto
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Takafumi Miyake
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
| | - Akira Tochio
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
- Department of Nephrology, Japanese Red Cross Society Wakayama Medical Center, Japan
| | - Akiko Mii
- Department of Nephrology, Nippon Medical School, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Japan
| | | | - Eri Muso
- Division of Nephrology and Dialysis, Kitano Hospital, Japan
- Department of Food and Nutrition, Faculty of Contemporary Home Economics, Kyoto Kacho University, Japan
| | - Motoko Yanagita
- Department of Nephrology, Kyoto University Graduate School of Medicine, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Japan
| |
Collapse
|
5
|
Nihei S, Ikeda T, Aoki T, Murasato F, Yaegashi M, Asahi K, Kudo K. Plasma endothelin-1 may predict bevacizumab-induced proteinuria in patients with colorectal cancer. Cancer Chemother Pharmacol 2023; 91:427-434. [PMID: 37036487 DOI: 10.1007/s00280-023-04532-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/30/2023] [Indexed: 04/11/2023]
Abstract
PURPOSE Proteinuria is one of the most common adverse events leading to the discontinuation of bevacizumab therapy. We analyzed plasma ET-1 levels as an indicator of renal endothelial dysfunction in colorectal cancer patients, to determine the utility of plasma ET-1 for identification of patients at high risk of proteinuria when treated with bevacizumab. METHODS Patients (n = 40) were recruited from an outpatient chemotherapy center between December 2020 and January 2022. Blood samples for plasma ET-1 levels were collected before treatment with bevacizumab (baseline), and after treatment for 3 and 6 months, and plasma ET-1 was determined by ELISA. Proteinuria was evaluated based on CTCAE v5.0 using urine protein-creatinine ratio instead of 24-h urine protein. RESULTS Plasma ET-1 levels at baseline were significantly higher in the group with grade ≥ 2 proteinuria than in the non-proteinuria group (p = 0.019). After adjusting for age, systolic and diastolic blood pressure, and hypertension following bevacizumab, plasma ET-1 levels at baseline were found to be an independent predictor of development of grade ≥ 2 proteinuria (OR = 17.8, 95% CI 1.42-223, and p = 0.026). Receiver operating characteristic curve analysis indicated an optimal cut-off value of the plasma ET-1 level of 1.19 pg/mL for predicting grade ≥ 2 proteinuria, with a sensitivity of 80.0% and specificity of 73.3%. CONCLUSION In conclusion, higher plasma ET-1 levels before treatment might increase the risk of proteinuria in colorectal cancer patients treated with bevacizumab. This might have important implications in the early detection of the risk of proteinuria.
Collapse
Affiliation(s)
- Satoru Nihei
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3695, Japan.
- Division of Clinical Pharmaceutics and Pharmacy Practice, Department of Clinical Pharmacy, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan.
| | - Tatsuki Ikeda
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3695, Japan
| | - Tomohiko Aoki
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3695, Japan
| | - Futa Murasato
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3695, Japan
| | - Mizunori Yaegashi
- Department of Surgery, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| | - Koichi Asahi
- Division of Nephrology and Hypertension, Department of Internal Medicine, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| | - Kenzo Kudo
- Department of Pharmacy, Iwate Medical University Hospital, 2-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3695, Japan
- Division of Clinical Pharmaceutics and Pharmacy Practice, Department of Clinical Pharmacy, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-Cho, Shiwa-Gun, Iwate, 028-3694, Japan
| |
Collapse
|
6
|
Abstract
Pre-eclampsia is a life-threatening disease of pregnancy unique to humans and a leading cause of maternal and neonatal morbidity and mortality. Women who survive pre-eclampsia have reduced life expectancy, with increased risks of stroke, cardiovascular disease and diabetes, while babies from a pre-eclamptic pregnancy have increased risks of preterm birth, perinatal death and neurodevelopmental disability and cardiovascular and metabolic disease later in life. Pre-eclampsia is a complex multisystem disease, diagnosed by sudden-onset hypertension (>20 weeks of gestation) and at least one other associated complication, including proteinuria, maternal organ dysfunction or uteroplacental dysfunction. Pre-eclampsia is found only when a placenta is or was recently present and is classified as preterm (delivery <37 weeks of gestation), term (delivery ≥37 weeks of gestation) and postpartum pre-eclampsia. The maternal syndrome of pre-eclampsia is driven by a dysfunctional placenta, which releases factors into maternal blood causing systemic inflammation and widespread maternal endothelial dysfunction. Available treatments target maternal hypertension and seizures, but the only 'cure' for pre-eclampsia is delivery of the dysfunctional placenta and baby, often prematurely. Despite decades of research, the aetiology of pre-eclampsia, particularly of term and postpartum pre-eclampsia, remains poorly defined. Significant advances have been made in the prediction and prevention of preterm pre-eclampsia, which is predicted in early pregnancy through combined screening and is prevented with daily low-dose aspirin, starting before 16 weeks of gestation. By contrast, the prediction of term and postpartum pre-eclampsia is limited and there are no preventive treatments. Future research must investigate the pathogenesis of pre-eclampsia, in particular of term and postpartum pre-eclampsia, and evaluate new prognostic tests and treatments in adequately powered clinical trials.
Collapse
|
7
|
Mohandes S, Doke T, Hu H, Mukhi D, Dhillon P, Susztak K. Molecular pathways that drive diabetic kidney disease. J Clin Invest 2023; 133:165654. [PMID: 36787250 PMCID: PMC9927939 DOI: 10.1172/jci165654] [Citation(s) in RCA: 110] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Kidney disease is a major driver of mortality among patients with diabetes and diabetic kidney disease (DKD) is responsible for close to half of all chronic kidney disease cases. DKD usually develops in a genetically susceptible individual as a result of poor metabolic (glycemic) control. Molecular and genetic studies indicate the key role of podocytes and endothelial cells in driving albuminuria and early kidney disease in diabetes. Proximal tubule changes show a strong association with the glomerular filtration rate. Hyperglycemia represents a key cellular stress in the kidney by altering cellular metabolism in endothelial cells and podocytes and by imposing an excess workload requiring energy and oxygen for proximal tubule cells. Changes in metabolism induce early adaptive cellular hypertrophy and reorganization of the actin cytoskeleton. Later, mitochondrial defects contribute to increased oxidative stress and activation of inflammatory pathways, causing progressive kidney function decline and fibrosis. Blockade of the renin-angiotensin system or the sodium-glucose cotransporter is associated with cellular protection and slowing kidney function decline. Newly identified molecular pathways could provide the basis for the development of much-needed novel therapeutics.
Collapse
Affiliation(s)
- Samer Mohandes
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hailong Hu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Chung EYM, Badve SV, Heerspink HJL, Wong MG. Endothelin receptor antagonists in kidney protection for diabetic kidney disease and beyond? Nephrology (Carlton) 2023; 28:97-108. [PMID: 36350038 PMCID: PMC10100079 DOI: 10.1111/nep.14130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
The burden of chronic kidney disease is increasing worldwide, largely due to the increasing global prevalence of diabetes mellitus and hypertension. While renin angiotensin system inhibitors and sodium-glucose cotransporter two inhibitors are the management cornerstone for reducing kidney and cardiovascular complications in patients with diabetic and non-diabetic kidney disease (DKD), they are partially effective and further treatments are needed to prevent the progression to kidney failure. Endothelin receptor antagonism represent a potential additional therapeutic option due to its beneficial effect on pathophysiological processes involved in progressive kidney disease including proteinuria, which are independently associated with progression of kidney disease. This review discusses the biological mechanisms of endothelin receptor antagonists (ERA) in kidney protection, the efficacy and safety of ERA in randomised controlled trials reporting on kidney outcomes, and its potential future use in both diabetic and non-DKDs.
Collapse
Affiliation(s)
- Edmund Y M Chung
- Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Sunil V Badve
- Renal and Metabolic Division, The George Institute for Global Health, University of New South Wales, Newtown, New South Wales, Australia.,Department of Renal Medicine, St George Hospital, Kogarah, New South Wales, Australia
| | - Hiddo J L Heerspink
- Renal and Metabolic Division, The George Institute for Global Health, University of New South Wales, Newtown, New South Wales, Australia.,Department of Clinical Pharmacoy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Muh Geot Wong
- Department of Renal Medicine, Concord Repatriation General Hospital, University of Sydney, Concord, New South Wales, Australia
| |
Collapse
|
9
|
Dines V, Suvakov S, Kattah A, Vermunt J, Narang K, Jayachandran M, Abou Hassan C, Norby AM, Garovic VD. Preeclampsia and the Kidney: Pathophysiology and Clinical Implications. Compr Physiol 2023; 13:4231-4267. [PMID: 36715282 DOI: 10.1002/cphy.c210051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preeclampsia and other hypertensive disorders of pregnancy are major contributors to maternal morbidity and mortality worldwide. This group of disorders includes chronic hypertension, gestational hypertension, preeclampsia, preeclampsia superimposed on chronic hypertension, and eclampsia. The body undergoes important physiological changes during pregnancy to allow for normal placental and fetal development. Several mechanisms have been proposed that may lead to preeclampsia, including abnormal placentation and placental hypoxia, impaired angiogenesis, excessive pro-inflammatory response, immune system imbalance, abnormalities of cellular senescence, alterations in regulation and activity of angiotensin II, and oxidative stress, ultimately resulting in upregulation of multiple mediators of endothelial cell dysfunction leading to maternal disease. The clinical implications of preeclampsia are significant as there are important short-term and long-term health consequences for those affected. Preeclampsia leads to increased risk of preterm delivery and increased morbidity and mortality of both the developing fetus and mother. Preeclampsia also commonly leads to acute kidney injury, and women who experience preeclampsia or another hypertensive disorder of pregnancy are at increased lifetime risk of chronic kidney disease and cardiovascular disease. An understanding of normal pregnancy physiology and the pathophysiology of preeclampsia is essential to develop novel treatment approaches and manage patients with preeclampsia and hypertensive disorders of pregnancy. © 2023 American Physiological Society. Compr Physiol 13:4231-4267, 2023.
Collapse
Affiliation(s)
- Virginia Dines
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jane Vermunt
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Kavita Narang
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Alexander M Norby
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.,Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
10
|
Otero González A. [NEPHROTIC SYNDROME AND PREGNANCY]. HIPERTENSION Y RIESGO VASCULAR 2023; 40:1-4. [PMID: 36906347 DOI: 10.1016/j.hipert.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 03/11/2023]
|
11
|
Bevacizumab Increases Endothelin-1 Production via Forkhead Box Protein O1 in Human Glomerular Microvascular Endothelial Cells In Vitro. Int J Nephrol 2021; 2021:8381115. [PMID: 34912580 PMCID: PMC8668358 DOI: 10.1155/2021/8381115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Molecular mechanisms underlying the nephrotoxicity associated with bevacizumab are unclear. Endothelin-1 (ET-1) is involved in podocyte injury and proteinuria, and its level increases in most cases of kidney disorders. Forkhead box protein O1 (FoxO1), a transcription factor, is a major determinant of ET-1 promoter activation and is regulated by protein kinase B (Akt) phosphorylation-dependent nuclear exclusion. We evaluated the effect of bevacizumab on ET-1 production in human glomerular microvascular endothelial cells (hGECs). We analyzed the changes in the mRNA and protein levels of ET-1 in hGECs treated with bevacizumab using real-time reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Changes in the protein levels and phosphorylation status of Akt and FoxO1 in hGECs treated with bevacizumab were analyzed by western blotting. After cell lysis, FoxO1 protein was isolated from the cytoplasmic and nuclear fractions. We also investigated the effects of AS1842856 (a FoxO1 inhibitor) on bevacizumab-induced ET-1 production. Bevacizumab significantly and dose-dependently increased the mRNA and protein levels of ET-1 in hGECs (p < 0.05). Bevacizumab treatment also led to a decrease in phosphorylated Akt protein levels. Inhibition of Akt activity by LY294002 promoted ET-1 production. Bevacizumab also induced an increase in FoxO1 protein levels in the nucleus. Inhibition of FoxO1 activity by AS1842856 resulted in decreased ET-1 levels in bevacizumab-treated hGECs. ET-1 axis activation, Akt inactivation, and FoxO1 nuclear localization are the molecular mechanisms underlying bevacizumab-induced nephrotoxicity. Therefore, inhibition of renal ET-1 production could be a promising approach to protect against or treat bevacizumab-induced nephrotoxicity.
Collapse
|
12
|
Mirabito Colafella KM, Neves KB, Montezano AC, Garrelds IM, van Veghel R, de Vries R, Uijl E, Baelde HJ, van den Meiracker AH, Touyz RM, Danser AHJ, Versmissen J. Selective ETA vs. dual ETA/B receptor blockade for the prevention of sunitinib-induced hypertension and albuminuria in WKY rats. Cardiovasc Res 2021; 116:1779-1790. [PMID: 31593221 DOI: 10.1093/cvr/cvz260] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/23/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS Although effective in preventing tumour growth, angiogenesis inhibitors cause off-target effects including cardiovascular toxicity and renal injury, most likely via endothelin (ET)-1 up-regulation. ET-1 via stimulation of the ETA receptor has pro-hypertensive actions whereas stimulation of the ETB receptor can elicit both pro- or anti-hypertensive effects. In this study, our aim was to determine the efficacy of selective ETA vs. dual ETA/B receptor blockade for the prevention of angiogenesis inhibitor-induced hypertension and albuminuria. METHODS AND RESULTS Male Wistar Kyoto (WKY) rats were treated with vehicle, sunitinib (angiogenesis inhibitor; 14 mg/kg/day) alone or in combination with macitentan (ETA/B receptor antagonist; 30 mg/kg/day) or sitaxentan (selective ETA receptor antagonist; 30 or 100 mg/kg/day) for 8 days. Compared with vehicle, sunitinib treatment caused a rapid and sustained increase in mean arterial pressure of ∼25 mmHg. Co-treatment with macitentan or sitaxentan abolished the pressor response to sunitinib. Sunitinib did not induce endothelial dysfunction. However, it was associated with increased aortic, mesenteric, and renal oxidative stress, an effect that was absent in mesenteric arteries of the macitentan and sitaxentan co-treated groups. Albuminuria was greater in the sunitinib- than vehicle-treated group. Co-treatment with sitaxentan, but not macitentan, prevented this increase in albuminuria. Sunitinib treatment increased circulating and urinary prostacyclin levels and had no effect on thromboxane levels. These increases in prostacyclin were blunted by co-treatment with sitaxentan. CONCLUSIONS Our results demonstrate that both selective ETA and dual ETA/B receptor antagonism prevents sunitinib-induced hypertension, whereas sunitinib-induced albuminuria was only prevented by selective ETA receptor antagonism. In addition, our results uncover a role for prostacyclin in the development of these effects. In conclusion, selective ETA receptor antagonism is sufficient for the prevention of sunitinib-induced hypertension and renal injury.
Collapse
Affiliation(s)
- Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Department of Physiology, Biomedicine Discovery Institute, Monash University, 26 Innovation Walk, Melbourne, VIC 3800, Australia
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Karla B Neves
- Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Augusto C Montezano
- Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ingrid M Garrelds
- Cardiovascular Disease Program, Department of Physiology, Biomedicine Discovery Institute, Monash University, 26 Innovation Walk, Melbourne, VIC 3800, Australia
| | - Richard van Veghel
- Cardiovascular Disease Program, Department of Physiology, Biomedicine Discovery Institute, Monash University, 26 Innovation Walk, Melbourne, VIC 3800, Australia
| | - René de Vries
- Cardiovascular Disease Program, Department of Physiology, Biomedicine Discovery Institute, Monash University, 26 Innovation Walk, Melbourne, VIC 3800, Australia
| | - Estrellita Uijl
- Cardiovascular Disease Program, Department of Physiology, Biomedicine Discovery Institute, Monash University, 26 Innovation Walk, Melbourne, VIC 3800, Australia
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anton H van den Meiracker
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Rhian M Touyz
- Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - A H Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Jorie Versmissen
- Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
13
|
Daehn IS, Duffield JS. The glomerular filtration barrier: a structural target for novel kidney therapies. Nat Rev Drug Discov 2021; 20:770-788. [PMID: 34262140 PMCID: PMC8278373 DOI: 10.1038/s41573-021-00242-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 12/19/2022]
Abstract
Loss of normal kidney function affects more than 10% of the population and contributes to morbidity and mortality. Kidney diseases are currently treated with immunosuppressive agents, antihypertensives and diuretics with partial but limited success. Most kidney disease is characterized by breakdown of the glomerular filtration barrier (GFB). Specialized podocyte cells maintain the GFB, and structure-function experiments and studies of intercellular communication between the podocytes and other GFB cells, combined with advances from genetics and genomics, have laid the groundwork for a new generation of therapies that directly intervene at the GFB. These include inhibitors of apolipoprotein L1 (APOL1), short transient receptor potential channels (TRPCs), soluble fms-like tyrosine kinase 1 (sFLT1; also known as soluble vascular endothelial growth factor receptor 1), roundabout homologue 2 (ROBO2), endothelin receptor A, soluble urokinase plasminogen activator surface receptor (suPAR) and substrate intermediates for coenzyme Q10 (CoQ10). These molecular targets converge on two key components of GFB biology: mitochondrial function and the actin-myosin contractile machinery. This Review discusses therapies and developments focused on maintaining GFB integrity, and the emerging questions in this evolving field.
Collapse
Affiliation(s)
- Ilse S Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jeremy S Duffield
- Research and Development, Prime Medicine, Cambridge, MA, USA. .,Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
14
|
Extracellular Vesicles Derived from Endothelial Progenitor Cells Protect Human Glomerular Endothelial Cells and Podocytes from Complement- and Cytokine-Mediated Injury. Cells 2021; 10:cells10071675. [PMID: 34359843 PMCID: PMC8304261 DOI: 10.3390/cells10071675] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022] Open
Abstract
Glomerulonephritis are renal inflammatory processes characterized by increased permeability of the Glomerular Filtration Barrier (GFB) with consequent hematuria and proteinuria. Glomerular endothelial cells (GEC) and podocytes are part of the GFB and contribute to the maintenance of its structural and functional integrity through the release of paracrine mediators. Activation of the complement cascade and pro-inflammatory cytokines (CK) such as Tumor Necrosis Factor α (TNF-α) and Interleukin-6 (IL-6) can alter GFB function, causing acute glomerular injury and progression toward chronic kidney disease. Endothelial Progenitor Cells (EPC) are bone-marrow-derived hematopoietic stem cells circulating in peripheral blood and able to induce angiogenesis and to repair injured endothelium by releasing paracrine mediators including Extracellular Vesicles (EVs), microparticles involved in intercellular communication by transferring proteins, lipids, and genetic material (mRNA, microRNA, lncRNA) to target cells. We have previously demonstrated that EPC-derived EVs activate an angiogenic program in quiescent endothelial cells and renoprotection in different experimental models. The aim of the present study was to evaluate in vitro the protective effect of EPC-derived EVs on GECs and podocytes cultured in detrimental conditions with CKs (TNF-α/IL-6) and the complement protein C5a. EVs were internalized in both GECs and podocytes mainly through a L-selectin-based mechanism. In GECs, EVs enhanced the formation of capillary-like structures and cell migration by modulating gene expression and inducing the release of growth factors such as VEGF-A and HGF. In the presence of CKs, and C5a, EPC-derived EVs protected GECs from apoptosis by decreasing oxidative stress and prevented leukocyte adhesion by inhibiting the expression of adhesion molecules (ICAM-1, VCAM-1, E-selectin). On podocytes, EVs inhibited apoptosis and prevented nephrin shedding induced by CKs and C5a. In a co-culture model of GECs/podocytes that mimicked GFB, EPC-derived EVs protected cell function and permeselectivity from inflammatory-mediated damage. Moreover, RNase pre-treatment of EVs abrogated their protective effects, suggesting the crucial role of RNA transfer from EVs to damaged glomerular cells. In conclusion, EPC-derived EVs preserved GFB integrity from complement- and cytokine-induced damage, suggesting their potential role as therapeutic agents for drug-resistant glomerulonephritis.
Collapse
|
15
|
Djenoune L, Tomar R, Dorison A, Ghobrial I, Schenk H, Hegermann J, Beverly-Staggs L, Hidalgo-Gonzalez A, Little MH, Drummond IA. Autonomous Calcium Signaling in Human and Zebrafish Podocytes Controls Kidney Filtration Barrier Morphogenesis. J Am Soc Nephrol 2021; 32:1697-1712. [PMID: 33911000 PMCID: PMC8425667 DOI: 10.1681/asn.2020101525] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/12/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Podocytes are critical to maintaining the glomerular filtration barrier, and mutations in nephrotic syndrome genes are known to affect podocyte calcium signaling. However, the role of calcium signaling during podocyte development remains unknown. METHODS We undertook live imaging of calcium signaling in developing podocytes, using zebrafish larvae and human kidney organoids. To evaluate calcium signaling during development and in response to channel blockers and genetic defects, the calcium biosensor GCaMP6s was expressed in zebrafish podocytes. We used electron microscopy to evaluate filtration barrier formation in zebrafish, and Fluo-4 to detect calcium signals in differentiating podocytes in human kidney organoids. RESULTS Immature zebrafish podocytes (2.5 days postfertilization) generated calcium transients that correlated with interactions with forming glomerular capillaries. Calcium transients persisted until 4 days postfertilization, and were absent after glomerular barrier formation was complete. We detected similar calcium transients in maturing human organoid glomeruli, suggesting a conserved mechanism. In both models, inhibitors of SERCA or IP3 receptor calcium-release channels blocked calcium transients in podocytes, whereas lanthanum was ineffective, indicating the calcium source is from intracellular podocyte endoplasmic-reticulum stores. Calcium transients were not affected by blocking heartbeat or by blocking development of endothelium or endoderm, and they persisted in isolated glomeruli, suggesting podocyte-autonomous calcium release. Inhibition of expression of phospholipase C-γ1, but not nephrin or phospholipase C-ε1, led to significantly decreased calcium activity. Finally, blocking calcium release affected glomerular shape and podocyte foot process formation, supporting the critical role of calcium signaling in glomerular morphogenesis. CONCLUSIONS These findings establish podocyte cell-autonomous calcium signaling as a prominent and evolutionarily conserved feature of podocyte differentiation and demonstrate its requirement for podocyte foot process formation.
Collapse
Affiliation(s)
- Lydia Djenoune
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Ritu Tomar
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Aude Dorison
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Irene Ghobrial
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany,Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Lynne Beverly-Staggs
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| | | | - Melissa H. Little
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Victoria, Australia
| | - Iain A. Drummond
- Davis Center for Regenerative Biology and Aging, Mount Desert Island Biological Laboratory, Bar Harbor, Maine
| |
Collapse
|
16
|
Ebefors K, Lassén E, Anandakrishnan N, Azeloglu EU, Daehn IS. Modeling the Glomerular Filtration Barrier and Intercellular Crosstalk. Front Physiol 2021; 12:689083. [PMID: 34149462 PMCID: PMC8206562 DOI: 10.3389/fphys.2021.689083] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
The glomerulus is a compact cluster of capillaries responsible for blood filtration and initiating urine production in the renal nephrons. A trilaminar structure in the capillary wall forms the glomerular filtration barrier (GFB), composed of glycocalyx-enriched and fenestrated endothelial cells adhering to the glomerular basement membrane and specialized visceral epithelial cells, podocytes, forming the outermost layer with a molecular slit diaphragm between their interdigitating foot processes. The unique dynamic and selective nature of blood filtration to produce urine requires the functionality of each of the GFB components, and hence, mimicking the glomerular filter in vitro has been challenging, though critical for various research applications and drug screening. Research efforts in the past few years have transformed our understanding of the structure and multifaceted roles of the cells and their intricate crosstalk in development and disease pathogenesis. In this review, we present a new wave of technologies that include glomerulus-on-a-chip, three-dimensional microfluidic models, and organoids all promising to improve our understanding of glomerular biology and to enable the development of GFB-targeted therapies. Here, we also outline the challenges and the opportunities of these emerging biomimetic systems that aim to recapitulate the complex glomerular filter, and the evolving perspectives on the sophisticated repertoire of cellular signaling that comprise the glomerular milieu.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
17
|
Endothelin-targeted new treatments for proteinuric and inflammatory glomerular diseases: focus on the added value to anti-renin-angiotensin system inhibition. Pediatr Nephrol 2021; 36:763-775. [PMID: 32185491 DOI: 10.1007/s00467-020-04518-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/06/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease (CKD) is the main cause of end-stage renal disease worldwide arising as a frequent complication of diabetes, obesity, and hypertension. Current therapeutic options, mainly based of inhibition of the renin-angiotensin system (RAS), provide imperfect renoprotection if started at an advanced phase of the disease, and treatments that show or even reverse the progression of CKD are needed. The endothelin (ET) system contributes to the normal renal physiology; however, robust evidence suggests a key role of ET-1 and its cognate receptors, in the progression of CKD. The effectiveness of ET receptor antagonists in ameliorating renal hemodynamics and fibrosis has been largely demonstrated in different experimental models. A significant antiproteinuric effect of ET receptor antagonists has been found in diabetic and non-diabetic CKD patients even on top of RAS blockade, and emerging evidence from ongoing clinical trials highlights their beneficial effects on a wide range of kidney disorders.
Collapse
|
18
|
Iampietro C, Bellucci L, Arcolino FO, Arigoni M, Alessandri L, Gomez Y, Papadimitriou E, Calogero RA, Cocchi E, Van Den Heuvel L, Levtchenko E, Bussolati B. Molecular and functional characterization of urine-derived podocytes from patients with Alport syndrome. J Pathol 2021; 252:88-100. [PMID: 32652570 PMCID: PMC7589231 DOI: 10.1002/path.5496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/25/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022]
Abstract
Alport syndrome (AS) is a genetic disorder involving mutations in the genes encoding collagen IV α3, α4 or α5 chains, resulting in the impairment of glomerular basement membrane. Podocytes are responsible for production and correct assembly of collagen IV isoforms; however, data on the phenotypic characteristics of human AS podocytes and their functional alterations are currently limited. The evident loss of viable podocytes into the urine of patients with active glomerular disease enables their isolation in a non‐invasive way. Here we isolated, immortalized, and subcloned podocytes from the urine of three different AS patients for molecular and functional characterization. AS podocytes expressed a typical podocyte signature and showed a collagen IV profile reflecting each patient's mutation. Furthermore, RNA‐sequencing analysis revealed 348 genes differentially expressed in AS podocytes compared with control podocytes. Gene Ontology analysis underlined the enrichment in genes involved in cell motility, adhesion, survival, and angiogenesis. In parallel, AS podocytes displayed reduced motility. Finally, a functional permeability assay, using a podocyte–glomerular endothelial cell co‐culture system, was established and AS podocyte co‐cultures showed a significantly higher permeability of albumin compared to control podocyte co‐cultures, in both static and dynamic conditions under continuous perfusion. In conclusion, our data provide a molecular characterization of immortalized AS podocytes, highlighting alterations in several biological processes related to extracellular matrix remodelling. Moreover, we have established an in vitro model to reproduce the altered podocyte permeability observed in patients with AS. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland..
Collapse
Affiliation(s)
- Corinne Iampietro
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Linda Bellucci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Fanny O Arcolino
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, University of Leuven, Leuven, Belgium
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luca Alessandri
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Yonathan Gomez
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Elli Papadimitriou
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Raffaele A Calogero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Enrico Cocchi
- Department of Pediatric Nephrology, University of Torino, Torino, Italy.,Division of Nephrology and Center for Precision Medicine and Genomics, Department of Medicine, Columbia University, New York, NY, USA
| | - Lambertus Van Den Heuvel
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, University of Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Laboratory of Pediatric Nephrology, Department of Development & Regeneration, University of Leuven, Leuven, Belgium.,Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| |
Collapse
|
19
|
Xie H, Wang H, Wu Q, Peng J, Huang H, Wang Y, Huang M, Jiang W, Yang Y, Zhang X, Zhang J, Zhu Q. Endothelin-1/Endothelin Receptor Type A-Angiopoietins/Tie-2 Pathway in Regulating the Cross Talk Between Glomerular Endothelial Cells and Podocytes in Trichloroethylene-Induced Renal Immune Injury. J Inflamm Res 2021; 14:761-776. [PMID: 33727850 PMCID: PMC7955787 DOI: 10.2147/jir.s301104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction This study aimed to investigate the mechanism in regulating the cross talk between glomerular endothelial cells and podocytes in “occupational medicamentosa-like dermatitis induced by trichloroethylene (OMLDT)” patients. Methods Totally 6 OMLDT patients, 18 controls, and 102 BALB/c female mice were involved in this study. Patient’s serum endothelin-1 (ET-1), angiopoietin-1 (Ang-1) and angiopoietin-2 (Ang-2), blood urea nitrogen (BUN), and podocalyxin (PCX) were detected. All the mice were used to establish the trichloroethylene (TCE) sensitized mouse model. Transmission electron microscope results were used to reflect renal glomerulus injury. Protein levels were detected by Western blot. Ang-1/Ang-2 gene level was reflected by RT-PCR. Cell apoptosis level was detected by using TUNEL assay kit. Results We found that in OMLDT patients, ET-1, Ang-2, BUN, and PCX were highly expressed but Ang-1 was inhibited. In TCE sensitized positive mouse, the downregulation of Ang-1, pTie-2 and the upregulation of Ang-2 were mediated by ET-1/ETAR but not ET-1/ETBR. The promotor of apoptosis proteins was downregulated and the inhibitor of apoptosis proteins was upregulated by treating with BQ123. Discussion ET-1/ETAR-Angs/Tie-2 pathway mediated the cross talk between glomerular endothelial cells and podocytes. BQ123 can alleviate glomerulus immune injury.
Collapse
Affiliation(s)
- Haibo Xie
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People's Republic of China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, People's Republic of China
| | - Hui Wang
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People's Republic of China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, People's Republic of China
| | - Qifeng Wu
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong Province, People's Republic of China
| | - Jiale Peng
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Hua Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Yican Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Meng Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Wei Jiang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Yi Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Xuesong Zhang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Jiaxiang Zhang
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, People's Republic of China.,Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui Province, People's Republic of China
| | - Qixing Zhu
- Department of Dermatology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People's Republic of China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, People's Republic of China
| |
Collapse
|
20
|
Li G, Wei W, Suo L, Zhang C, Yu H, Liu H, Guo Q, Zhen X, Yu Y. Low-Dose Aspirin Prevents Kidney Damage in LPS-Induced Preeclampsia by Inhibiting the WNT5A and NF-κB Signaling Pathways. Front Endocrinol (Lausanne) 2021; 12:639592. [PMID: 33790866 PMCID: PMC8006287 DOI: 10.3389/fendo.2021.639592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/15/2021] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is a serious pregnancy-related disease, and patients usually present with a high inflammatory response. Previous studies have suggested that aspirin (ASP) may have a role in alleviating the pathogenesis of preeclampsia. However, whether ASP can improve kidney damage and the mechanism for improving it is currently unclear. Here we optimized a lipopolysaccharide (LPS)-induced PE mouse model to identify the role of ASP in renal protection. We found that ASP treatment ameliorated LPS-induced renal failure and pathological changes, the tubular injury was significantly attenuated by ASP. Administration of ASP decreased the renal expression of pro-inflammatory factors, resulting in reduced kidney inflammation. The number of GALECTIN-3-positive cells was reduced, and the up-regulation of IL-6 and TNF-α was decreased. In addition, ASP also suppressed renal cell apoptosis and oxidative stress. An in vitro study indicated that ASP relieved LPS-induced HK-2 cell damage by inhibiting WNT5A/NF-κB signaling. Collectively, our data suggest that ASP is a useful therapeutic option for PE-related kidney injury.
Collapse
Affiliation(s)
- Guanlin Li
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Wei Wei
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Lingge Suo
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Chun Zhang
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing, China
| | - Haiyan Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Qing Guo
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Xiumei Zhen
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
21
|
Sol M, Kamps JAAM, van den Born J, van den Heuvel MC, van der Vlag J, Krenning G, Hillebrands JL. Glomerular Endothelial Cells as Instigators of Glomerular Sclerotic Diseases. Front Pharmacol 2020; 11:573557. [PMID: 33123011 PMCID: PMC7573930 DOI: 10.3389/fphar.2020.573557] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Glomerular endothelial cell (GEnC) dysfunction is important in the pathogenesis of glomerular sclerotic diseases, including Focal Segmental Glomerulosclerosis (FSGS) and overt diabetic nephropathy (DN). GEnCs form the first cellular barrier in direct contact with cells and factors circulating in the blood. Disturbances in these circulating factors can induce GEnC dysfunction. GEnC dysfunction occurs in early stages of FSGS and DN, and is characterized by a compromised endothelial glycocalyx, an inflammatory phenotype, mitochondrial damage and oxidative stress, aberrant cell signaling, and endothelial-to-mesenchymal transition (EndMT). GEnCs are in an interdependent relationship with podocytes and mesangial cells, which involves bidirectional cross-talk via intercellular signaling. Given that GEnC behavior directly influences podocyte function, it is conceivable that GEnC dysfunction may culminate in podocyte damage, proteinuria, subsequent mesangial activation, and ultimately glomerulosclerosis. Indeed, GEnC dysfunction is sufficient to cause podocyte injury, proteinuria and activation of mesangial cells. Aberrant gene expression patterns largely contribute to GEnC dysfunction and epigenetic changes seem to be involved in causing aberrant transcription. This review summarizes literature that uncovers the importance of cross-talk between GEnCs and podocytes, and GEnCs and mesangial cells in the context of the development of FSGS and DN, and the potential use of GEnCs as efficacious cellular target to pharmacologically halt development and progression of DN and FSGS.
Collapse
Affiliation(s)
- Marloes Sol
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jan A A M Kamps
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jacob van den Born
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marius C van den Heuvel
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Guido Krenning
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
22
|
Rush BM, Bondi CD, Stocker SD, Barry KM, Small SA, Ong J, Jobbagy S, Stolz DB, Bastacky SI, Chartoumpekis DV, Kensler TW, Tan RJ. Genetic or pharmacologic Nrf2 activation increases proteinuria in chronic kidney disease in mice. Kidney Int 2020; 99:102-116. [PMID: 32818518 DOI: 10.1016/j.kint.2020.07.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) pathway upregulates key cellular defenses. Clinical trials are utilizing pharmacologic Nrf2 inducers such as bardoxolone methyl to treat chronic kidney disease, but Nrf2 activation has been linked to a paradoxical increase in proteinuria. To understand this effect, we examined genetically engineered mice with elevated Nrf2 signaling due to reduced expression of the Nrf2 inhibitor, Kelch-like ECH-associated protein 1 (Keap1). These Keap1FA/FA mice lacked baseline proteinuria but exhibited increased proteinuria in experimental models evoked by adriamycin, angiotensin II, or protein overload. After injury, Keap1FA/FA mice had increased glomerulosclerosis, nephrin disruption and shedding, podocyte injury, foot process effacement, and interstitial fibrosis. Keap1FA/FA mice also had higher daytime blood pressures and lower heart rates measured by radiotelemetry. Conversely, Nrf2 knockout mice were protected from proteinuria. We also examined the pharmacologic Nrf2 inducer CDDO-Im. Compared to angiotensin II alone, the combination of angiotensin II and CDDO-Im significantly increased proteinuria, a phenomenon not observed in Nrf2 knockout mice. This effect was not accompanied by additional increases in blood pressure. Finally, Nrf2 was found to be upregulated in the glomeruli of patients with focal segmental glomerulosclerosis, diabetic nephropathy, fibrillary glomerulonephritis, and membranous nephropathy. Thus, our studies demonstrate that Nrf2 induction in mice may exacerbate proteinuria in chronic kidney disease.
Collapse
Affiliation(s)
- Brittney M Rush
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Corry D Bondi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sean D Stocker
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kacie M Barry
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah A Small
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jason Ong
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Soma Jobbagy
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Donna B Stolz
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dionysios V Chartoumpekis
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas W Kensler
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA; Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Roderick J Tan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
23
|
Kwiatkowska E, Stefańska K, Zieliński M, Sakowska J, Jankowiak M, Trzonkowski P, Marek-Trzonkowska N, Kwiatkowski S. Podocytes-The Most Vulnerable Renal Cells in Preeclampsia. Int J Mol Sci 2020; 21:ijms21145051. [PMID: 32708979 PMCID: PMC7403979 DOI: 10.3390/ijms21145051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022] Open
Abstract
Preeclampsia (PE) is a disorder that affects 3–5% of normal pregnancies. It was believed for a long time that the kidney, similarly to all vessels in the whole system, only sustained endothelial damage. The current knowledge gives rise to a presumption that the main role in the development of proteinuria is played by damage to the podocytes and their slit diaphragm. The podocyte damage mechanism in preeclampsia is connected to free VEGF and nitric oxide (NO) deficiency, and an increased concentration of endothelin-1 and oxidative stress. From national cohort studies, we know that women who had preeclampsia in at least one pregnancy carried five times the risk of developing end-stage renal disease (ESRD) when compared to women with physiological pregnancies. The focal segmental glomerulosclerosis (FSGS) is the dominant histopathological lesion in women with a history of PE. The kidney’s podocytes are not subject to replacement or proliferation. Podocyte depletion exceeding 20% resulted in FSGS, which is a reason for the later development of ESRD. In this review, we present the mechanism of kidney (especially podocytes) injury in preeclampsia. We try to explain how this damage affects further changes in the morphology and function of the kidneys after pregnancy.
Collapse
Affiliation(s)
- Ewa Kwiatkowska
- Clinical Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, 80-210 Gdańsk, Poland
- Correspondence:
| | - Maciej Zieliński
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, 80-210 Gdańsk, Poland; (M.Z.); (J.S.); (M.J.); (P.T.)
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science Cancer Immunology Group, University of Gdansk, 80-822 Gdańsk, Poland;
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Sebastian Kwiatkowski
- Department of Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW We focus on the current understanding of preeclampsia (PE) in order to examine how it mediates glomerular injury and affects the course of glomerulonephritis (GNs). In addition, this review discusses the role of GNs on the development of PE. RECENT FINDINGS In PE, the dysfunctional utero-placental perfusion causes the release into the mother's circulation of anti-angiogenic substances, leading to systemic endotheliosis. In preeclamptic patients, the imbalance between pro- and anti-angiogenic factors is responsible for the kidney injury, and PE may reveal a silent pre-existent GN or may induce the development of the disease. Moreover, in women with chronic kidney disease (CKD), it could accelerate the disease progression. In any case, GNs compromise renal function, making the kidney less responsive to physiological changes that occur during pregnancy and, at the same time, cause maternal vascular inflammation, representing a risk factor for PE development. Although a bidirectional correlation between GNs and PE has been demonstrated, the data are limited, and further large studies are warranted. Close collaboration between a multidisciplinary team of obstetricians and nephrologists is essential to establish the correct diagnosis and safely manage these vulnerable women and their fetuses.
Collapse
|
25
|
Yuan M, Tan Y, Wang Y, Wang SX, Yu F, Zhao MH. The associations of endothelial and podocyte injury in proliferative lupus nephritis: from observational analysis to in vitro study. Lupus 2019; 28:347-358. [PMID: 30755145 DOI: 10.1177/0961203319828509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Our study aims to evaluate the endothelial cell-podocyte crosstalk in proliferative lupus nephritis (LN). The semi-quantification scores of glomerular endothelial cell injury and the foot process width (FPW) were processed in 110 proliferative LN patients. Podocytes were stimulated with LN-derived IgG. Glomerular endothelial cells were treated with podocyte-conditioned medium (PCM), and then podocytes were incubated with endothelial cell-conditioned medium (ECM). The levels of vascular endothelial growth factor-A (VEGF-A) in PCM and endothelin-1 in ECM were analyzed, and the injury of podocyte and glomerular endothelial cells were further evaluated. The pathological score of glomerular endothelial cell injury was correlated with FPW in LN complicated with thrombotic microangiopathy. In vitro study showed the following: 1. Stimulation of podocytes by IgG from LN led to decline in the expression of nephrin with cytoskeleton rearrangement, and reduction of VEGF-A levels. 2. Exposure of glomerular endothelial cells to PCM incubated with LN-derived IgG (PCM-LN) induced more endothelin-1 secretion and disruption of intercellular tight junction. 3. Exposure of podocytes to ECM stimulated with PCM-LN could induce cytoskeleton redistribution with decrease of nephrin. In conclusion, the pathological glomerular endothelial cell lesions were associated with FPW and the VEGF-endothelin-1 system might play a critical role in the endothelial cell-podocyte crosstalk in LN.
Collapse
Affiliation(s)
- M Yuan
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Y Tan
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - Y Wang
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - S X Wang
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China
| | - F Yu
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China.,5 Department of Nephrology, Peking University International Hospital, Beijing, People's Republic of China
| | - M H Zhao
- 1 Renal Division, Department of Medicine, Peking University First Hospital, Beijing, People's Republic of China.,2 Institute of Nephrology, Peking University, Beijing, People's Republic of China.,3 Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, People's Republic of China.,4 Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Ministry of Education of China, Beijing, People's Republic of China.,6 Peking-Tsinghua Center for Life Sciences, Beijing, People's Republic of China
| |
Collapse
|
26
|
Alghamdi TA, Batchu SN, Hadden MJ, Yerra VG, Liu Y, Bowskill BB, Advani SL, Geldenhuys L, Siddiqi FS, Majumder S, Advani A. Histone H3 Serine 10 Phosphorylation Facilitates Endothelial Activation in Diabetic Kidney Disease. Diabetes 2018; 67:2668-2681. [PMID: 30213824 DOI: 10.2337/db18-0124] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/30/2018] [Indexed: 11/13/2022]
Abstract
The posttranslational histone modifications that epigenetically affect gene transcription extend beyond conventionally studied methylation and acetylation patterns. By examining the means by which podocytes influence the glomerular endothelial phenotype, we identified a role for phosphorylation of histone H3 on serine residue 10 (phospho-histone H3Ser10) in mediating endothelial activation in diabetes. Culture media conditioned by podocytes exposed to high glucose caused glomerular endothelial vascular cell adhesion protein 1 (VCAM-1) upregulation and was enriched for the chemokine CCL2. A neutralizing anti-CCL2 antibody prevented VCAM-1 upregulation in cultured glomerular endothelial cells, and knockout of the CCL2 receptor CCR2 diminished glomerular VCAM-1 upregulation in diabetic mice. CCL2/CCR2 signaling induced glomerular endothelial VCAM-1 upregulation through a pathway regulated by p38 mitogen-activated protein kinase, mitogen- and stress-activated protein kinases 1/2 (MSK1/2), and phosphorylation of H3Ser10, whereas MSK1/2 inhibition decreased H3Ser10 phosphorylation at the VCAM1 promoter. Finally, increased phospho-histone H3Ser10 levels were observed in the kidneys of diabetic endothelial nitric oxide synthase knockout mice and in the glomeruli of humans with diabetic kidney disease. These findings demonstrate the influence that histone protein phosphorylation may have on gene activation in diabetic kidney disease. Histone protein phosphorylation should be borne in mind when considering epigenetic targets amenable to therapeutic manipulation in diabetes.
Collapse
Affiliation(s)
- Tamadher A Alghamdi
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Sri N Batchu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Mitchell J Hadden
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Veera Ganesh Yerra
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Youan Liu
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Bridgit B Bowskill
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | | | - Ferhan S Siddiqi
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Syamantak Majumder
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Zhao B, Han H, Zhen J, Yang X, Shang J, Xu L, Wang R. CD80 and CTLA-4 as diagnostic and prognostic markers in adult-onset minimal change disease: a retrospective study. PeerJ 2018; 6:e5400. [PMID: 30083478 PMCID: PMC6078067 DOI: 10.7717/peerj.5400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022] Open
Abstract
Background Minimal change disease (MCD) is a form of idiopathic nephrotic syndrome. Compared to children, adult-onset MCD patients are reported to have delayed responses to glucocorticoid treatment. Several studies of children have suggested detecting urinary CD80 levels to diagnose MCD. There are no effective diagnostic methods to distinguish steroid-sensitive MCD from steroid-resistant MCD unless treatments are used. Methods In a total of 55 patients with biopsy-proven MCD and 26 patients with biopsy-proven idiopathic membranous nephropathy, CD80 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) levels in serum, urine and renal tissue were analyzed. Results Steroid-sensitive MCD patients in remission had lower urinary CD80 levels and higher CTLA-4 levels than patients in relapse (156.65 ± 24.62 vs 1066.40 ± 176.76 ng/g creatinine; p < 0.0001), (728.73 ± 89.93 vs 151.70 ± 27.01 ng/g creatinine; p < 0.0001). For MCD patients in relapse, mean urinary CD80 level was higher, and CTLA-4 level was lower for those who were steroid-sensitive than those who were steroid-resistant (1066.40 ± 176.76 vs. 203.78 ± 30.65 ng/g creatinine; p < 0.0001), but the mean urinary CTLA-4 level was lower (151.70 ± 27.01 vs. 457.83 ± 99.45 ng/g creatinine; p < 0.0001). CD80 expression in glomeruli was a sensitive marker to diagnose MCD. The absent or minimal expression of CTLA-4 in glomeruli could distinguish steroid-sensitive MCD from steroid-resistant MCD. Conclusions Glucocorticoid treatment may result in complete remission for only MCD patients with strongly positive CD80 expression and negative CTLA-4 expression in glomeruli, or higher urinary CD80 level and lower CTLA-4 level.
Collapse
Affiliation(s)
- Bing Zhao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Hui Han
- Department of Intensive Care Unit, Shandong University Qilu Hospital, Jinan, China
| | - Junhui Zhen
- School of Medicine, Shandong University, Jinan, China
| | - Xiaowei Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Xu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
28
|
Armaly Z, Jadaon JE, Jabbour A, Abassi ZA. Preeclampsia: Novel Mechanisms and Potential Therapeutic Approaches. Front Physiol 2018; 9:973. [PMID: 30090069 PMCID: PMC6068263 DOI: 10.3389/fphys.2018.00973] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/02/2018] [Indexed: 01/04/2023] Open
Abstract
Preeclampsia is a serious complication of pregnancy where it affects 5–8% of all pregnancies. It increases the morbidity and mortality of both the fetus and pregnant woman, especially in developing countries. It deleteriously affects several vital organs, including the kidneys, liver, brain, and lung. Although, the pathogenesis of preeclampsia has not yet been fully understood, growing evidence suggests that aberrations in the angiogenic factors levels and coagulopathy are responsible for the clinical manifestations of the disease. The common nominator of tissue damage of all these target organs is endothelial injury, which impedes their normal function. At the renal level, glomerular endothelial injury leads to the development of maternal proteinuria. Actually, peripheral vasoconstriction secondary to maternal systemic inflammation and endothelial cell activation is sufficient for the development of preeclampsia-induced hypertension. Similarly, preeclampsia can cause hepatic and neurologic dysfunction due to vascular damage and/or hypertension. Obviously, preeclampsia adversely affects various organs, however it is not yet clear whether pre-eclampsia per se adversely affects various organs or whether it exposes underlying genetic predispositions to cardiovascular disease that manifest in later life. The current review summarizes recent development in the pathogenesis of preeclampsia with special focus on novel diagnostic biomarkers and their relevance to potential therapeutic options for this disease state. Specifically, the review highlights the renal manifestations of the disease with emphasis on the involvement of angiogenic factors in vascular injury and on how restoration of the angiogenic balance affects renal and cardiovascular outcome of Preeclamptic women.
Collapse
Affiliation(s)
- Zaher Armaly
- Department of Nephrology, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Jimmy E Jadaon
- Department of Obstetrics and Gynecology, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel.,Laboratory Medicine, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Adel Jabbour
- Laboratory Medicine, EMMS Nazareth Hospital, Galilee Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Zaid A Abassi
- Department of Physiology, The Ruth and Burce Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel.,Department of Laboratory Medicine, Rambam Health Campus, Haifa, Israel
| |
Collapse
|
29
|
Bakrania BA, Spradley FT, Satchell SC, Stec DE, Rimoldi JM, Gadepalli RSV, Granger JP. Heme oxygenase-1 is a potent inhibitor of placental ischemia-mediated endothelin-1 production in cultured human glomerular endothelial cells. Am J Physiol Regul Integr Comp Physiol 2018; 314:R427-R432. [PMID: 29212810 PMCID: PMC5899255 DOI: 10.1152/ajpregu.00370.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/07/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder of maternal hypertension and reduced renal hemodynamics linked to reduced endothelial function. Placental ischemia is thought to be the culprit of this disease, as it causes the release of factors like tumor necrosis factor (TNF)-α that induce vascular endothelin-1 (ET-1) production. Interestingly, placental ischemia-induced hypertension in rats [reduced uterine perfusion pressure (RUPP) model] is abolished by ETA receptor blockade, suggesting a critical role for ET-1. Although it has been found that systemic induction of heme oxygenase (HO)-1 is associated with reduced ET-1 production and attenuated hypertension, it is unclear whether HO-1 directly modulates the increased ET-1 response to placental factors. We tested the hypothesis that HO-1 or its metabolites inhibit ET-1 production in human glomerular endothelial cells induced by serum of RUPP rats or TNF-α. Serum (5%) from RUPP hypertensive (mean arterial blood pressure 119 ± 9 mmHg) vs. normotensive pregnant (NP, 101 ± 6 mmHg, P < 0.001) rats increased ET-1 production (RUPP 168.8 ± 18.1 pg/ml, NP 80.3 ± 22.7 pg/ml, P < 0.001, n = 12/group). HO-1 induction [25 µM cobalt photoporphyrin (CoPP)] abolished RUPP serum-induced ET-1 production (1.6 ± 0.8 pg/ml, P < 0.001), whereas bilirubin (10 µM) significantly attenuated ET-1 release (125.3 ± 5.2 pg/ml, P = 0.005). Furthermore, TNF-α-induced ET-1 production (TNF-α 31.0 ± 8.4 vs. untreated 7.5 ± 0.4 pg/ml, P < 0.001) was reduced by CoPP (1.5 ± 0.8 pg/ml, P < 0.001) and bilirubin (10.5 ± 4.3 pg/ml, P < 0.001). These results suggest that circulating factors released during placental ischemia target the maternal glomerular endothelium to increase ET-1, and that pharmacological induction of HO-1 or bilirubin could be a treatment strategy to block this prohypertensive pathway in preeclampsia.
Collapse
Affiliation(s)
- Bhavisha A Bakrania
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Frank T Spradley
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Surgery, University of Mississippi Medical Center , Jackson, Mississippi
| | - Simon C Satchell
- School of Clinical Sciences, University of Bristol , Bristol , United Kingdom
| | - David E Stec
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - John M Rimoldi
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Rama S V Gadepalli
- Department of Biomolecular Sciences, University of Mississippi , Oxford, Mississippi
| | - Joey P Granger
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
30
|
van de Lest NA, Zandbergen M, IJpelaar DHT, Wolterbeek R, Bruijn JA, Bajema IM, Scharpfenecker M. Nephrin Loss Can Be Used to Predict Remission and Long-term Renal Outcome in Patients With Minimal Change Disease. Kidney Int Rep 2017; 3:168-177. [PMID: 29340328 PMCID: PMC5762955 DOI: 10.1016/j.ekir.2017.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/30/2017] [Accepted: 09/18/2017] [Indexed: 11/16/2022] Open
Abstract
Introduction Minimal change disease is a common cause of nephrotic syndrome. In general, patients with minimal change disease respond to corticosteroids and have excellent long-term renal survival. However, some patients have less favorable outcome. These patients are often thought to have progressed to focal segmental glomerulosclerosis. We previously reported that a segmental loss of podocyte markers is present before the development of focal segmental glomerulosclerosis in a rat model. Here, we investigated whether loss of podocyte marker nephrin can serve as a biomarker for predicting poor outcome in patients with minimal change disease. Methods We obtained 47 kidney biopsy samples from patients diagnosed with minimal change disease and stained sections with periodic acid−Schiff and for nephrin. Nephrin loss was scored by 2 independent researchers who were blinded to clinical outcome. Clinical data were collected retrospectively, and nephrin loss was correlated with clinical follow-up data. Results Nephrin loss was present in 34% of the biopsy samples. During follow-up, patients with nephrin loss achieved remission less frequently (61%) compared to patients without (96%) (P = 0.002). Moreover, 5-year eGFR was lower in the patients with renal nephrin loss. The risk of eGFR decreasing to < 60 ml/min per 1.73m2 increased with each percentage of glomeruli with nephrin loss (hazard ratio = 1.044, 95% confidence interval = 1.02−1.07). Conclusion These results indicate that nephrin loss in patients with minimal change disease can help predict both remission and long-term renal outcome.
Collapse
Affiliation(s)
- Nina A van de Lest
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Malu Zandbergen
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Daphne H T IJpelaar
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan A Bruijn
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ingeborg M Bajema
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
31
|
Kasztan M, Fox BM, Speed JS, De Miguel C, Gohar EY, Townes TM, Kutlar A, Pollock JS, Pollock DM. Long-Term Endothelin-A Receptor Antagonism Provides Robust Renal Protection in Humanized Sickle Cell Disease Mice. J Am Soc Nephrol 2017; 28:2443-2458. [PMID: 28348063 DOI: 10.1681/asn.2016070711] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/13/2017] [Indexed: 11/03/2022] Open
Abstract
Sickle cell disease (SCD)-associated nephropathy is a major source of morbidity and mortality in patients because of the lack of efficacious treatments targeting renal manifestations of the disease. Here, we describe a long-term treatment strategy with the selective endothelin-A receptor (ETA) antagonist, ambrisentan, designed to interfere with the development of nephropathy in a humanized mouse model of SCD. Ambrisentan preserved GFR at the level of nondisease controls and prevented the development of proteinuria, albuminuria, and nephrinuria. Microscopy studies demonstrated prevention of podocyte loss and structural alterations, the absence of vascular congestion, and attenuation of glomerulosclerosis in treated mice. Studies in isolated glomeruli showed that treatment reduced inflammation and oxidative stress. At the level of renal tubules, ambrisentan treatment prevented the increased excretion of urinary tubular injury biomarkers. Additionally, the treatment strategy prevented tubular brush border loss, diminished tubular iron deposition, blocked the development of interstitial fibrosis, and prevented immune cell infiltration. Furthermore, the prevention of albuminuria in treated mice was associated with preservation of cortical megalin expression. In a separate series of identical experiments, combined ETA and ETB receptor antagonism provided only some of the protection observed with ambrisentan, highlighting the importance of exclusively targeting the ETA receptor in SCD. Our results demonstrate that ambrisentan treatment provides robust protection from diverse renal pathologies in SCD mice, and suggest that long-term ETA receptor antagonism may provide a strategy for the prevention of renal complications of SCD.
Collapse
Affiliation(s)
| | - Brandon M Fox
- Cardio-Renal Physiology and Medicine, Department of Medicine, and
| | - Joshua S Speed
- Cardio-Renal Physiology and Medicine, Department of Medicine, and
| | - Carmen De Miguel
- Cardio-Renal Physiology and Medicine, Department of Medicine, and
| | - Eman Y Gohar
- Cardio-Renal Physiology and Medicine, Department of Medicine, and
| | - Tim M Townes
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama; and
| | | | - Jennifer S Pollock
- Cardio-Renal Physiology and Medicine, Department of Medicine, and.,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - David M Pollock
- Cardio-Renal Physiology and Medicine, Department of Medicine, and .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
32
|
Tien WS, Chen JH, Wu KP. SheddomeDB: the ectodomain shedding database for membrane-bound shed markers. BMC Bioinformatics 2017; 18:42. [PMID: 28361715 PMCID: PMC5374707 DOI: 10.1186/s12859-017-1465-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A number of membrane-anchored proteins are known to be released from cell surface via ectodomain shedding. The cleavage and release of membrane proteins has been shown to modulate various cellular processes and disease pathologies. Numerous studies revealed that cell membrane molecules of diverse functional groups are subjected to proteolytic cleavage, and the released soluble form of proteins may modulate various signaling processes. Therefore, in addition to the secreted protein markers that undergo secretion through the secretory pathway, the shed membrane proteins may comprise an additional resource of noninvasive and accessible biomarkers. In this context, identifying the membrane-bound proteins that will be shed has become important in the discovery of clinically noninvasive biomarkers. Nevertheless, a data repository for biological and clinical researchers to review the shedding information, which is experimentally validated, for membrane-bound protein shed markers is still lacking. RESULTS In this study, the database SheddomeDB was developed to integrate publicly available data of the shed membrane proteins. A comprehensive literature survey was performed to collect the membrane proteins that were verified to be cleaved or released in the supernatant by immunological-based validation experiments. From 436 studies on shedding, 401 validated shed membrane proteins were included, among which 199 shed membrane proteins have not been annotated or validated yet by existing cleavage databases. SheddomeDB attempted to provide a comprehensive shedding report, including the regulation of shedding machinery and the related function or diseases involved in the shedding events. In addition, our published tool ShedP was embedded into SheddomeDB to support researchers for predicting the shedding event on unknown or unrecorded membrane proteins. CONCLUSIONS To the best of our knowledge, SheddomeDB is the first database for the identification of experimentally validated shed membrane proteins and currently may provide the most number of membrane proteins for reviewing the shedding information. The database included membrane-bound shed markers associated with numerous cellular processes and diseases, and some of these markers are potential novel markers because they are not annotated or validated yet in other databases. SheddomeDB may provide a useful resource for discovering membrane-bound shed markers. The interactive web of SheddomeDB is publicly available at http://bal.ym.edu.tw/SheddomeDB/ .
Collapse
Affiliation(s)
- Wei-Sheng Tien
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.,Bioinformatics Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 115, Taiwan
| | - Jun-Hong Chen
- Department of Computer Science, National Taipei University of Education, Taipei, 106, Taiwan
| | - Kun-Pin Wu
- Institute of Biomedical Informatics, National Yang Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
33
|
Jung YJ, Cho HY, Cho S, Kim YH, Jeon JD, Kim YJ, Lee S, Park J, Kim HY, Park YW, Kwon JY. The Level of Serum and Urinary Nephrin in Normal Pregnancy and Pregnancy with Subsequent Preeclampsia. Yonsei Med J 2017; 58:401-406. [PMID: 28120572 PMCID: PMC5290021 DOI: 10.3349/ymj.2017.58.2.401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/21/2016] [Accepted: 09/02/2016] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate serum and urinary nephrin levels of normal pregnancy to establish a standard reference value and to compare them with patients who subsequently developed preeclampsia (PE). MATERIALS AND METHODS In this prospective study, 117 healthy singleton pregnancies were enrolled between 6 to 20 weeks of gestation at 2 participating medical centers during October 2010 to March 2012. Urine and serum samples were collected at the time of enrollment, each trimester, and at 4 to 6 weeks postpartum. Enzyme-linked immunosorbent assay for nephrin was performed and samples from patients who subsequently developed PE were compared to the normal patients. RESULTS Of 117 patients initially enrolled, 99 patients delivered at the study centers and of those patients, 12 (12.1%) developed PE at a median gestational age of 34⁺⁴ weeks (range 29⁺⁵-36⁺⁶). In the normal patients (n=68), serum nephrin level decreased and urinary nephrin level increased during the latter of pregnancy. In 12 patients who subsequently developed PE, a significant rise in the 3rd trimester serum and urinary nephrin levels, compared to the controls, was observed (p<0.001), and this increase occurred 9 days prior to the onset of clinical disease. CONCLUSION As the onset of PE was preceded by the rise in the serum and urinary nephrin in comparison to normal pregnancy, serum and urinary nephrin may be a useful predictive marker of PE.
Collapse
Affiliation(s)
- Yun Ji Jung
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hee Young Cho
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sihyun Cho
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Young Han Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Dong Jeon
- Department of Obstetrics and Gynecology, MizMedi Women's Hospital, Seoul, Korea
| | - Young Jin Kim
- Department of Bioanalysis, Seoul Medical Science Institute & Seoul Clinical Laboratories, Seoul, Korea
| | - Sanghoo Lee
- Department of Bioanalysis, Seoul Medical Science Institute & Seoul Clinical Laboratories, Seoul, Korea
| | - Jimyeong Park
- Department of Bioanalysis, Seoul Medical Science Institute & Seoul Clinical Laboratories, Seoul, Korea
| | - Ha Yan Kim
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Won Park
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ja Young Kwon
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
34
|
Kato T, Mizuno S. Nephron, Wilms' tumor-1 (WT1), and synaptopodin expression in developing podocytes of mice. Exp Anim 2017; 66:183-189. [PMID: 28179596 PMCID: PMC5543238 DOI: 10.1538/expanim.16-0101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Newborn mouse glomeruli are still immature with a morphological feature of an early
capillary loop stage, but infant mice do not manifest proteinuria. Little is known about
the molecular mechanism whereby infant mice are resistant to proteinuria. Nephrin and
synaptopodin are crucial for slit diaphragm and foot process (FP) formation for avoiding
proteinuria. Nephrin tyrosine phosphorylation means a transient biological signaling
required for FP repair or extension during nephrotic disease. Using an immunohistochemical
technique, we examined the natural course of nephrin, Wilms’ tumor-1 (WT1) and
synaptopodin at 16.5 days of embryonic age (E16.5d) and E19.5d, 7 days of post-neonatal
age (P7d) and P42d during renal development of mice. As a result, nephrin and synaptopodin
were detected at E19.5d in S-shaped bodies. WT1, a transcriptional factor for nephrin, was
detected in nucleus in podocyte-like cells in all stages. Nephrin tyrosine phosphorylation
was evident in glomeruli at P7d, and this was associated with an early-stage of FP
extension. Inversely, nephrin phosphorylation became faint at P42d, along with maturated
FP. Based on the present results, we suggest the sequential molecular mechanism to protect
growing mice from proteinuria: (i) WT1-induced nephrin production by podocytes in S-shaped
bodies at E19.5d; (ii) Synchronized induction of synaptopodin at the same period; and
(iii) FP extension is initiated at a milk-suckling stage under a nephrin
tyrosine-phosphorylated condition, while it is arrested at an adult stage, associated with
a loss of nephrin-based signaling.
Collapse
Affiliation(s)
- Takashi Kato
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Department of Pathology, Faculty of Medicine, Kindai University, 377-2 Ohno-higashi, Osaka-sayama, Osaka 589-8511, Japan
| | - Shinya Mizuno
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.,Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
35
|
Angiotensin II increases glomerular permeability by β-arrestin mediated nephrin endocytosis. Sci Rep 2016; 6:39513. [PMID: 28004760 PMCID: PMC5177899 DOI: 10.1038/srep39513] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 11/21/2016] [Indexed: 01/13/2023] Open
Abstract
Glomerular permeability and subsequent albuminuria are early clinical markers for glomerular injury in hypertensive nephropathy. Albuminuria predicts mortality and cardiovascular morbidity. AT1 receptor blockers protect from albuminuria, cardiovascular morbidity and mortality. A blood pressure independent, molecular mechanism for angiotensin II (Ang II) dependent albuminuria has long been postulated. Albuminuria results from a defective glomerular filter. Nephrin is a major structural component of the glomerular slit diaphragm and its endocytosis is mediated by β-arrestin2. Ang II stimulation increases nephrin-β-arrestin2 binding, nephrin endocytosis and glomerular permeability in mice. This Ang II effect is mediated by AT1-receptors. AT1-receptor mutants identified G-protein signaling to be essential for this Ang II effect. Gαq knockdown and phospholipase C inhibition block Ang II mediated enhanced nephrin endocytosis. Nephrin Y1217 is the critical residue controlling nephrin binding to β-arrestin under Ang II stimulation. Nephrin Y1217 also mediates cytoskeletal anchoring to actin via nck2. Ang II stimulation decreases nephrin nck2 binding. We conclude that Ang II weakens the structural integrity of the slit diaphragm by increased nephrin endocytosis and decreased nephrin binding to nck2, which leads to increased glomerular permeability. This novel molecular mechanism of Ang II supports the use of AT1-receptor blockers to prevent albuminuria even in normotensives.
Collapse
|
36
|
Collapsing glomerulopathy is common in the setting of thrombotic microangiopathy of the native kidney. Kidney Int 2016; 90:1321-1331. [DOI: 10.1016/j.kint.2016.07.021] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 12/11/2022]
|
37
|
van den Meiracker AH, Danser AHJ. Mechanisms of Hypertension and Renal Injury During Vascular Endothelial Growth Factor Signaling Inhibition. Hypertension 2016; 68:17-23. [PMID: 27185750 DOI: 10.1161/hypertensionaha.116.07618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Anton H van den Meiracker
- From the Division of Pharmacology and Cardiovascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | - A H Jan Danser
- From the Division of Pharmacology and Cardiovascular Medicine, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Abstract
The incidence of progressive kidney disease associated with diabetes continues to increase worldwide. Only partial renoprotection is achieved by current standard therapy with angiotensin-converting enzyme inhibitors and/or angiotensin-receptor blockers, increasing the need for novel therapeutic approaches. Experimental studies have provided evidence of a pathogenic role for endothelin-1 (ET-1) and its cognate receptors in the development and progression of diabetic nephropathy. ET-1, mainly through the activation of ETA receptor, contributes to renal cell injury, inflammation, and fibrosis. In animal models of type 1 and type 2 diabetes, ETA-selective antagonists have been shown to provide renoprotective effects, supplying the rationale for clinical trials in patients with diabetic nephropathy with ETA-receptor antagonists administered in addition to renin-angiotensin system blockade.
Collapse
Affiliation(s)
- Elena Gagliardini
- Unit of Advanced Microscopy, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Carlamaria Zoja
- Laboratory of Pathophysiology of Experimental Renal Disease and Interaction With Other Organ Systems, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Ariela Benigni
- Department of Molecular Medicine, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy.
| |
Collapse
|
39
|
Culshaw GJ, MacIntyre IM, Dhaun N, Webb DJ. Endothelin in nondiabetic chronic kidney disease: preclinical and clinical studies. Semin Nephrol 2016; 35:176-87. [PMID: 25966349 DOI: 10.1016/j.semnephrol.2015.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The incidence and prevalence of chronic kidney disease (CKD) is increasing. Despite current therapies, many patients with CKD have suboptimal blood pressure, ongoing proteinuria, and develop progressive renal dysfunction. Further therapeutic options therefore are required. Over the past 20 years the endothelin (ET) system has become a prime target. Experimental models have shown that ET-1, acting primarily via the endothelin-A receptor, plays an important role in the development of proteinuria, glomerular injury, fibrosis, and inflammation. Subsequent animal and early clinical studies using ET-receptor antagonists have suggested that theses therapies may slow renal disease progression primarily through blood pressure and proteinuria reduction. This review examines the current literature regarding the ET system in nondiabetic CKD.
Collapse
Affiliation(s)
- Geoff J Culshaw
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.
| | - Iain M MacIntyre
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Neeraj Dhaun
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - David J Webb
- University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| |
Collapse
|
40
|
Abstract
Endothelin-1 (ET-1) is a 21-amino acid peptide with mitogenic and powerful vasoconstricting properties. Under healthy conditions, ET-1 is expressed constitutively in all cells of the glomerulus and participates in homeostasis of glomerular structure and filtration function. Under disease conditions, increases in ET-1 are critically involved in initiating and maintaining glomerular inflammation, glomerular basement membrane hypertrophy, and injury of podocytes (visceral epithelial cells), thereby promoting proteinuria and glomerulosclerosis. Here, we review the role of ET-1 in the function of glomerular endothelial cells, visceral (podocytes) and parietal epithelial cells, mesangial cells, the glomerular basement membrane, stromal cells, inflammatory cells, and mesenchymal stem cells. We also discuss molecular mechanisms by which ET-1, predominantly through activation of the ETA receptor, contributes to injury to glomerular cells, and review preclinical and clinical evidence supporting its pathogenic role in glomerular injury in chronic renal disease. Finally, the therapeutic rationale for endothelin antagonists as a new class of antiproteinuric drugs is discussed.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zurich, Zurich, Switzerland.
| | - Andrey Sorokin
- Department of Medicine, Kidney Disease Center, Division of Nephrology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
41
|
Abstract
OBJECTIVES Nephrin is an integral part of podocytes that together with endothelial cells and the basement form the glomerular filtration barrier. Placental ischemia triggers a cascade of events that ultimately result in endothelial malfunction, hypertension, podocytopathy and fetal compromise. METHODS We review the literature to determine if urine nephrin measurements could serve as a useful biomarker to detect early podocyte injury in pre-eclampsia. RESULTS Our search identifies eight studies published to date. The findings of these studies demonstrate that urine nephrin excretion plays a critical role in the pathogenesis of proteinuria during pre-eclampsia and that this is a good indicator of glomerular injury. CONCLUSION There is thus an urgent need for a large multi-centre clinical study using standardized recruitment criteria to determine the full potential of this biomarker in clinical practice.
Collapse
Affiliation(s)
- Yogavijayan Kandasamy
- a Department of Neonatology , The Townsville Hospital , Douglas , Queensland , Australia .,b Mothers and Babies Research Centre, Hunter Medical Research Institute, John Hunter Hospital, The University of Newcastle , Callaghan , New South Wales , Australia .,c College of Public Health, Medical and Veterinary Sciences, The James Cook University , Townsville City , Queensland , Australia , and
| | - David Watson
- d Department of Obstetrics and Gynaecology , The Townsville Hospital , Douglas , Queensland , Australia
| | - Donna Rudd
- c College of Public Health, Medical and Veterinary Sciences, The James Cook University , Townsville City , Queensland , Australia , and
| |
Collapse
|
42
|
Yesildag B, Bock T, Herrmanns K, Wollscheid B, Stoffel M. Kin of IRRE-like Protein 2 Is a Phosphorylated Glycoprotein That Regulates Basal Insulin Secretion. J Biol Chem 2015; 290:25891-906. [PMID: 26324709 DOI: 10.1074/jbc.m115.684704] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Indexed: 12/17/2022] Open
Abstract
Direct interactions among pancreatic β-cells via cell surface proteins inhibit basal and enhance stimulated insulin secretion. Here, we functionally and biochemically characterized Kirrel2, an immunoglobulin superfamily protein with β-cell-specific expression in the pancreas. Our results show that Kirrel2 is a phosphorylated glycoprotein that co-localizes and interacts with the adherens junction proteins E-cadherin and β-catenin in MIN6 cells. We further demonstrate that the phosphosites Tyr(595-596) are functionally relevant for the regulation of Kirrel2 stability and localization. Analysis of the extracellular and intracellular domains of Kirrel2 revealed that it is cleaved and shed from MIN6 cells and that the remaining membrane spanning cytoplasmic domain is processed by γ-secretase complex. Kirrel2 knockdown with RNA interference in MIN6 cells and ablation of Kirrel2 from mice with genetic deletion resulted in increased basal insulin secretion from β-cells, with no immediate influence on stimulated insulin secretion, total insulin content, or whole body glucose metabolism. Our results show that in pancreatic β-cells Kirrel2 localizes to adherens junctions, is regulated by multiple post-translational events, including glycosylation, extracellular cleavage, and phosphorylation, and engages in the regulation of basal insulin secretion.
Collapse
Affiliation(s)
- Burcak Yesildag
- From the Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich
| | - Thomas Bock
- the Department of Health Sciences and Technology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology Zurich, Auguste-Piccard-Hof 1, 8093 Zurich, and
| | - Karolin Herrmanns
- From the Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich
| | - Bernd Wollscheid
- the Department of Health Sciences and Technology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology Zurich, Auguste-Piccard-Hof 1, 8093 Zurich, and
| | - Markus Stoffel
- From the Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zurich, the Department of Health Sciences and Technology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology Zurich, Auguste-Piccard-Hof 1, 8093 Zurich, and the Faculty of Medicine, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
43
|
Lankhorst S, Baelde HJ, Kappers MHW, Smedts FMM, Hansen A, Clahsen-van Groningen MC, Sleijfer S, Mathijssen RHJ, Danser AHJ, van den Meiracker AH. Greater Sensitivity of Blood Pressure Than Renal Toxicity to Tyrosine Kinase Receptor Inhibition With Sunitinib. Hypertension 2015. [PMID: 26195484 DOI: 10.1161/hypertensionaha.115.05435] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypertension and renal injury are off-target effects of sunitinib, a tyrosine kinase receptor inhibitor used for the treatment of various tumor types. Importantly, these untoward effects are accompanied by activation of the endothelin system. Here, we set up a study to explore the dose dependency of these side effects. Normotensive Wistar Kyoto rats were exposed to 3 different doses of sunitinib or vehicle. After 8 days, rats were euthanized. Telemetrically measured blood pressure rose dose dependently, from 13 to 30 mm Hg. Proteinuria was present at all doses, but a rise in cystatin C occurred only at the intermediate and high doses. Compared with vehicle circulating endothelin-1 increased dose dependently, whereas 24-hour urinary endothelin excretion decreased. Light and electron microscopy revealed glomerular endotheliosis and ischemia with the intermediate and high doses of sunitinib but completely absent histological abnormalities with the low dose. Podocyte number per glomerular circumference did not change. Glomerular nephrin, Neph1, podocin, and endothelin-converting enzyme gene expression were downregulated in a dose-dependent manner. We conclude that the sunitinib-induced rise in blood pressure requires lower doses than its induction of renal function impairment and that functional changes in glomerular filtration barrier contribute to the occurrence of proteinuria, given the lack of histopathologic changes with the low dose of sunitinib.
Collapse
Affiliation(s)
- Stephanie Lankhorst
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Hans J Baelde
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Mariëtte H W Kappers
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Frank M M Smedts
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Alastair Hansen
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Marian C Clahsen-van Groningen
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Stefan Sleijfer
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - Ron H J Mathijssen
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| | - A H Jan Danser
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.).
| | - Anton H van den Meiracker
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands (S.L., M.H.W.K., F.M.M.S., A.H.J.D., A.H.v.d.M.); Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands (H.J.B.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); Department of Pathology, Reinier de Graaf Gasthuis, Delft, The Netherlands (F.M.M.S.); Department of Pathology, Herlev University Hospital, Herlev, Denmark (A.H.); Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands (M.C.C.-v.G.); and Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus Medical Center, Rotterdam, The Netherlands (S.S., R.H.JM.)
| |
Collapse
|
44
|
Yun BH, Lee SM, Cho HY, Kim JY, Son GH, Kim YH, Park YW, Lim BJ, Kwon JY. Expression of nephrin in the human placenta and fetal membranes. Mol Med Rep 2015; 12:5116-20. [PMID: 26151763 DOI: 10.3892/mmr.2015.4044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 11/20/2014] [Indexed: 11/05/2022] Open
Abstract
Nephrin is the signature molecule in the podocyte of the glomerulus that forms the renal slit diaphragm, the main functional unit of the glomerulus. The present study focused on the expression of nephrin in the human placenta, which may also have a role in filtration and the maintenance of homeostasis in the kidneys. A total of nine placentas from normal healthy pregnant females at full term were investigated. Reverse transcription-quantitative polymerase chain reaction, western blotting and immunofluorescence were performed. The expression of nephrin mRNA was relatively increased in the chorion compared with that in the villi and the amnion. The nephrin gene was detected in the villous cytotrophoblast cells and the endothelium of the intravillous vessels. It was also present in the chorionic and amniotic membranous lining, with its distribution being particularly dense in the amniocytes. The identification of nephrin in the human placenta, particularly at the maternal‑fetal interface, provides a novel insight into the molecular basis of the selective permeability of the placental barrier, which requires further elucidation.
Collapse
Affiliation(s)
- Bo Hyon Yun
- Division of Maternal‑Fetal Medicine, Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Seung Mi Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 110‑799, Republic of Korea
| | - Hee Young Cho
- Division of Maternal‑Fetal Medicine, Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Ji Young Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Ga Hyun Son
- Department of Obstetrics and Gynecology, Kangnam Sacred Heart Hospital, Hallym University Medical Center, Seoul 150‑719, Republic of Korea
| | - Young Han Kim
- Division of Maternal‑Fetal Medicine, Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Yong Won Park
- Division of Maternal‑Fetal Medicine, Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| | - Beom Jin Lim
- Department of Pathology, Gangnam Severance Hospital, Yonsei University Health System, Seoul 135‑720, Republic of Korea
| | - Ja Young Kwon
- Division of Maternal‑Fetal Medicine, Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul 120‑752, Republic of Korea
| |
Collapse
|
45
|
From Glomerular Endothelium to Podocyte Pathobiology in Preeclampsia: a Paradigm Shift. Curr Hypertens Rep 2015; 17:54. [DOI: 10.1007/s11906-015-0566-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
46
|
Kwiatkowski S, Kwiatkowska E, Rzepka R, Kurkiewicz V, Mikołajek-Bedner W, Torbè A. Development of a focal segmental glomerulosclerosis after pregnancy complicated by preeclampsia: case report and review of literature. J Matern Fetal Neonatal Med 2015; 29:1566-9. [DOI: 10.3109/14767058.2015.1053865] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
47
|
Histamine receptor expression in human renal tubules: a comparative pharmacological evaluation. Inflamm Res 2015; 64:261-70. [PMID: 25725698 DOI: 10.1007/s00011-015-0807-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 02/08/2015] [Accepted: 02/19/2015] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE AND DESIGN The aim of this study is to evaluate the expression of the histamine receptors, particularly focusing on the H4R in human renal tubules. MATERIAL The ex vivo evaluation was carried on specimens from human renal cortex. Primary and immortalized tubular epithelial cells (TECs) and the HK-2 cell line were used as in vitro models. TREATMENT Cells were pretreated for 10 min with chlorpheniramine maleate 10 μM (H1R antagonist), ranitidine 10 µM (H2R antagonist), GSK189254 1 µM (H3R antagonist) or JNJ7777120 10 µM (H4R antagonist), and then exposed to histamine (3 pM-10 nM) for 30 min. METHODS The ex vivo evaluation on specimens from human renal cortex was performed by immunohistochemistry. The expression of histamine receptors on primary and immortalized TECs and the HK-2 cell line was evaluated at both gene (RT-PCR) and protein (immunocytofluorescence) levels. The pharmacological analysis was performed by TR-FRET measurements of second messenger (IP3 and cAMP) production induced by histamine with or without the selective antagonists. RESULTS Our data revealed the presence of all histamine receptors in human tubules; however, only TECs expressed all the receptors. Indeed, histamine elicited a sigmoid dose-response curve for IP3 production, shifted to the right by chlorpheniramine maleate, and elicited a double bell-shaped curve for cAMP production, partially suppressed by the selective H2R, H3R and H4R antagonists when each added alone, and completely ablated when combined together. CONCLUSIONS Herein, we report the identification of all four histamine receptors in human renal tubules.
Collapse
|
48
|
Heimlich JB, Speed JS, Bloom CJ, O'Connor PM, Pollock JS, Pollock DM. ET-1 increases reactive oxygen species following hypoxia and high-salt diet in the mouse glomerulus. Acta Physiol (Oxf) 2015; 213:722-30. [PMID: 25219340 DOI: 10.1111/apha.12397] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 09/04/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022]
Abstract
AIM This study was designed to determine whether ET-1 derived from endothelial cells contributes to oxidative stress in the glomerulus of mice subjected to a high-salt diet and/or hypoxia. METHODS C57BL6/J control mice or vascular endothelial cell ET-1 knockout (VEET KO) mice were subjected to 3-h exposure to hypoxia (8% O₂) and/or 2 weeks of high-salt diet (4% NaCl) prior to metabolic cage assessment of renal function and isolation of glomeruli for the determination of reactive oxygen species (ROS). RESULTS In control mice, hypoxia significantly increased urinary protein excretion during the initial 24 h, but only in animals on a high-salt diet. Hypoxia increased glomerular ET-1 mRNA expression in control, but not in vascular endothelial cell ET-1 knockout (VEET KO) mice. Under normoxic conditions, mice on a high-salt diet had approx. 150% higher glomerular ET-1 mRNA expression compared with a normal-salt diet (P < 0.05). High-salt diet administration significantly increased glomerular ROS production in flox control, but not in glomeruli isolated from VEET KO mice. In C57BL6/J mice, the ETA receptor-selective antagonist, ABT-627, significantly attenuated the increase in glomerular ROS production produced by high-salt diet. In addition, chronic infusion of C57BL6/J mice with a subpressor dose of ET-1 (osmotic pumps) significantly increased the levels of glomerular ROS that were prevented by ETA antagonist treatment. CONCLUSION These data suggest that both hypoxia and a high-salt diet increase glomerular ROS production via endothelial-derived ET-1-ETA receptor activation and provide a potential mechanism for ET-1-induced nephropathy.
Collapse
Affiliation(s)
- J. B. Heimlich
- Department of Physiology; Georgia Regents University; Augusta GA USA
| | - J. S. Speed
- Cardio-Renal Physiology and Medicine; Division of Nephrology; University of Alabama at Birmingham; Birmingham AL USA
| | - C. J. Bloom
- Department of Medicine; Georgia Regents University; Augusta GA USA
| | - P. M. O'Connor
- Department of Physiology; Georgia Regents University; Augusta GA USA
| | - J. S. Pollock
- Cardio-Renal Physiology and Medicine; Division of Nephrology; University of Alabama at Birmingham; Birmingham AL USA
| | - D. M. Pollock
- Cardio-Renal Physiology and Medicine; Division of Nephrology; University of Alabama at Birmingham; Birmingham AL USA
| |
Collapse
|
49
|
Kohan DE, Barton M. Endothelin and endothelin antagonists in chronic kidney disease. Kidney Int 2014; 86:896-904. [PMID: 24805108 PMCID: PMC4216619 DOI: 10.1038/ki.2014.143] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 01/31/2014] [Accepted: 02/06/2014] [Indexed: 01/10/2023]
Abstract
The incidence and prevalence of chronic kidney disease (CKD), with diabetes and hypertension accounting for the majority of cases, is on the rise, with up to 160 million individuals worldwide predicted to be affected by 2020. Given that current treatment options, primarily targeted at the renin-angiotensin system, only modestly slow down progression to end-stage renal disease, the urgent need for additional effective therapeutics is evident. Endothelin-1 (ET-1), largely through activation of endothelin A receptors, has been strongly implicated in renal cell injury, proteinuria, inflammation, and fibrosis leading to CKD. Endothelin receptor antagonists (ERAs) have been demonstrated to ameliorate or even reverse renal injury and/or fibrosis in experimental models of CKD, whereas clinical trials indicate a substantial antiproteinuric effect of ERAs in diabetic and nondiabetic CKD patients even on top of maximal renin-angiotensin system blockade. This review summarizes the role of ET in CKD pathogenesis and discusses the potential therapeutic benefit of targeting the ET system in CKD, with attention to the risks and benefits of such an approach.
Collapse
Affiliation(s)
- Donald E. Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, UT
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
50
|
Lankhorst S, Kappers MHW, van Esch JHM, Smedts FMM, Sleijfer S, Mathijssen RHJ, Baelde HJ, Danser AHJ, van den Meiracker AH. Treatment of hypertension and renal injury induced by the angiogenesis inhibitor sunitinib: preclinical study. Hypertension 2014; 64:1282-9. [PMID: 25185126 DOI: 10.1161/hypertensionaha.114.04187] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Common adverse effects of angiogenesis inhibition are hypertension and renal injury. To determine the most optimal way to prevent these adverse effects and to explore their interdependency, the following drugs were investigated in unrestrained Wistar Kyoto rats exposed to the angiogenesis inhibitor sunitinib: the dual endothelin receptor antagonist macitentan; the calcium channel blocker amlodipine; the angiotensin-converting enzyme inhibitor captopril; and the phosphodiesterase type 5 inhibitor sildenafil. Mean arterial pressure was monitored telemetrically. After 8 days, rats were euthanized and blood samples and kidneys were collected. In addition, 24-hour urine samples were collected. After sunitinib start, mean arterial pressure increased rapidly by ≈30 mm Hg. Coadministration of macitentan or amlodipine largely prevented this rise, whereas captopril or sildenafil did not. Macitentan, captopril, and sildenafil diminished the sunitinib-induced proteinuria and endothelinuria and glomerular intraepithelial protein deposition, whereas amlodipine did not. Changes in proteinuria and endothelinuria were unrelated. We conclude that in our experimental model, dual endothelin receptor antagonism and calcium channel blockade are suitable to prevent angiogenesis inhibition-induced hypertension, whereas dual endothelin receptor antagonism, angiotensin-converting enzyme inhibitor, and phosphodiesterase type 5 inhibition can prevent angiogenesis inhibition-induced proteinuria. Moreover, the variable response of hypertension and renal injury to different antihypertensive agents suggests that these side effects are, at least in part, unrelated.
Collapse
Affiliation(s)
- Stephanie Lankhorst
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Mariëtte H W Kappers
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Joep H M van Esch
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Frank M M Smedts
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Stefan Sleijfer
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Ron H J Mathijssen
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Hans J Baelde
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - A H Jan Danser
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.)
| | - Anton H van den Meiracker
- From the Division of Pharmacology and Vascular Medicine, Department of Internal Medicine (S.L., M.H.W.K., J.H.M.v.E., A.H.J.D., A.H.v.d.M.) and Department of Medical Oncology, Erasmus MC Cancer Institute (S.S., R.H.J.M.), Erasmus Medical Center, Rotterdam, The Netherlands; Department of Pathology, Reinier de Graaf Groep, Delft, The Netherlands (F.M.M.S.); Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands (M.H.W.K.); and Department of Pathology, Leiden University Medical Center, Leiden (H.J.B.).
| |
Collapse
|