1
|
Ren H, Mu C, Wang Y, Cheng Y, Hou Y, Li Y, Liu N, Yin Z, Xiong H, Chen Y, Yang T, Yu Y, Shen Y. Notch2 Inhibition and Kidney Cyst Growth in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2025:00001751-990000000-00518. [PMID: 39745791 DOI: 10.1681/asn.0000000592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Key Points
Notch2 activation promotes kidney cyst growth.Silencing Notch2 ameliorated cyst growth in mice with autosomal dominant polycystic kidney disease.
Background
Notch signaling, a conserved mechanism of cell-to-cell communication, plays a crucial role in regulating cellular processes, such as proliferation and differentiation, in a context-dependent manner. However, the specific contribution of Notch signaling to the progression of polycystic kidney disease (PKD) remains unclear.
Methods
We investigated the changes in Notch signaling activity (Notch1–4) in the kidneys of patients with autosomal dominant PKD (ADPKD) and two ADPKD mouse models (early and late onset). Multiple genetic and pharmacologic approaches were used to explore Notch2 signaling during kidney cyst formation in PKD.
Results
Notch2 expression was significantly increased in the kidney tissues of patients with ADPKD and ADPKD mice. Targeted expression of Notch2 intracellular domain in renal epithelial cells resulted in cyst formation and kidney failure in neonatal and adult mice. Mechanistically, Notch2/Hey2 signaling promoted renal epithelial cell proliferation by driving the expression of the E26 transformation–specific homologous factor (Ehf). Depletion of Ehf delayed Notch2 intracellular domain overexpression–induced cyst formation and kidney failure in mice. A gain-of-function mutation in exon 34 of NOTCH2 (c.6426dupT), which caused PKD in patients with Hajdu–Cheney syndrome, accelerated cell growth in cultured human renal epithelial cells by activating HEY2/EHF signaling. Finally, ablation of Notch2 or treatment of a kidney-targeting nanoparticle carrying the liposome/Notch2–small interfering RNA complex, significantly suppressed kidney cyst growth in early-onset ADPKD mice.
Conclusions
Notch2 signaling promoted kidney cyst growth, partially by upregulating Ehf expression.
Collapse
Affiliation(s)
- Huiwen Ren
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chengsen Mu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuhan Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Cheng
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yayan Hou
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yizhe Li
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Na Liu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhuming Yin
- Department of Breast Oncoplastic Surgery, Key Laboratory of Breast Cancer Prevention and Therapy, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Sino-Russian Joint Research Center for Oncoplastic Breast Surgery, Tianjin Medical University, Tianjin, China
| | - Hui Xiong
- Department of Urology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tianxin Yang
- Department of Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yujun Shen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Haihe Laboratory of Cell Ecosystem, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
2
|
Cornelius RJ, Maeoka Y, Shinde U, McCormick JA. Familial Hyperkalemic Hypertension. Compr Physiol 2024; 14:5839-5874. [PMID: 39699086 DOI: 10.1002/cphy.c240004] [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: 12/20/2024]
Abstract
The rare disease Familial Hyperkalemic Hypertension (FHHt) is caused by mutations in the genes encoding Cullin 3 (CUL3), Kelch-Like 3 (KLHL3), and two members of the With-No-Lysine [K] (WNK) kinase family, WNK1 and WNK4. In the kidney, these mutations ultimately cause hyperactivation of NCC along the renal distal convoluted tubule. Hypertension results from increased NaCl retention, and hyperkalemia by impaired K + secretion by downstream nephron segments. CUL3 and KLHL3 are now known to form a ubiquitin ligase complex that promotes proteasomal degradation of WNK kinases, which activate downstream kinases that phosphorylate and thus activate NCC. For CUL3, potent effects on the vasculature that contribute to the more severe hypertensive phenotype have also been identified. Here we outline the in vitro and in vivo studies that led to the discovery of the molecular pathways regulating NCC and vascular tone, and how FHHt-causing mutations disrupt these pathways. Potential mechanisms for variability in disease severity related to differential effects of each mutation on the kidney and vasculature are described, and other possible effects of the mutant proteins beyond the kidney and vasculature are explored. © 2024 American Physiological Society. Compr Physiol 14:5839-5874, 2024.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Yujiro Maeoka
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Ujwal Shinde
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
3
|
Rodan AR. With No Lysine (K) Kinases and Sodium Transporter Function in Solute Exchange with Implications for BP Regulation as Elucidated through Drosophila. KIDNEY360 2024; 5:1553-1562. [PMID: 39186374 PMCID: PMC11556937 DOI: 10.34067/kid.0000000000000564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Like other multicellular organisms, the fruit fly Drosophila melanogaster must maintain homeostasis of the internal milieu, including the maintenance of constant ion and water concentrations. In mammals, the with no lysine (K) (WNK)-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade is an important regulator of epithelial ion transport in the kidney. This pathway regulates SLC12 family cotransporters, including sodium-potassium-2-chloride, sodium chloride, and potassium chloride cotransporters. The WNK-Ste20-proline/alanine rich kinase/oxidative stress response 1 kinase cascade also regulates epithelial ion transport via regulation of the Drosophila sodium-potassium-2-chloride cotransporter in the Malpighian tubule, the renal epithelium of the fly. Studies in Drosophila have contributed to the understanding of multiple regulators of WNK pathway signaling, including intracellular chloride and potassium, the scaffold protein Mo25, hypertonic stress, hydrostatic pressure, and macromolecular crowding. These will be discussed together, with implications for mammalian kidney function and BP control.
Collapse
Affiliation(s)
- Aylin R Rodan
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah; Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, Utah; Department of Human Genetics, University of Utah, Salt Lake City, Utah; and Medical Service, Veterans Affairs Salt Lake City Health Care System, Salt Lake City, Utah
| |
Collapse
|
4
|
Maeoka Y, Bradford T, Su XT, Sharma A, Yang CL, Ellison DH, McCormick JA, Cornelius RJ. Distal convoluted tubule-specific disruption of the COP9 signalosome but not its regulatory target cullin 3 causes tubular injury. Am J Physiol Renal Physiol 2024; 327:F667-F682. [PMID: 39205661 PMCID: PMC11483082 DOI: 10.1152/ajprenal.00138.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
The disease familial hyperkalemic hypertension (FHHt; also known as Gordon syndrome) is caused by aberrant accumulation of with-no-lysine kinase (WNK4) activating the NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) of the kidney. Mutations in cullin 3 (CUL3) cause FHHt by disrupting interaction with the deneddylase COP9 signalosome (CSN). Deletion of Cul3 or Jab1 (the catalytically active CSN subunit) along the entire nephron causes a partial FHHt phenotype with activation of the WNK4-STE20/SPS1-related proline/alanine-rich kinase (SPAK)-NCC pathway. However, progressive kidney injury likely prevents hypertension, hyperkalemia, and hyperchloremic metabolic acidosis associated with FHHt. We hypothesized that DCT-specific deletion would more closely model the disease. We used Slc12a3-Cre-ERT2 mice to delete Cul3 (DCT-Cul3-/-) or Jab1 (DCT-Jab1-/-) only in the DCT and examined the mice after short- and long-term deletion. Short-term DCT-specific knockout of both Cul3 and Jab1 mice caused elevated WNK4, pSPAKS373, and pNCCT53 abundance. However, neither model demonstrated changes in plasma K+, Cl-, or total CO2, even though no injury was present. Long-term DCT-Jab1-/- mice showed significantly lower NCC and parvalbumin abundance and a higher abundance of kidney injury molecule-1, a marker of proximal tubule injury. No injury or reduction in NCC or parvalbumin was observed in long-term DCT-Cul3-/- mice. In summary, the prevention of injury outside the DCT did not lead to a complete FHHt phenotype despite activation of the WNK4-SPAK-NCC pathway, possibly due to insufficient NCC activation. Chronically, only DCT-Jab1-/- mice developed tubule injury and atrophy of the DCT, suggesting a direct JAB1 effect or dysregulation of other cullins as mechanisms for injury.NEW & NOTEWORTHY CUL3 degrades WNK4, which prevents activation of NCC in the DCT. CSN regulation of CUL3 is impaired in the disease FHHt, causing accumulation of WNK4. Short-term DCT-specific disruption of CUL3 or the CSN in mice resulted in activation of the WNK4-SPAK-NCC pathway but not hyperkalemic metabolic acidosis found in FHHt. Tubule injury was observed only after long-term CSN disruption. The data suggest that disruption of other cullins may be the cause for the injury.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Tanner Bradford
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
- LeDucq Transatlantic Network of Excellence, Boston, Massachusetts, United States
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
5
|
Ferdaus MZ, Terker AS, Koumangoye RB, Al-Qusairi L, Welling PA, Delpire E. Deletion of KS-WNK1 promotes NCC activation by increasing WNK1/4 abundance. Am J Physiol Renal Physiol 2024; 327:F373-F385. [PMID: 38961847 PMCID: PMC11460338 DOI: 10.1152/ajprenal.00101.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Dietary potassium deficiency causes stimulation of sodium reabsorption leading to an increased risk in blood pressure elevation. The distal convoluted tubule (DCT) is the main rheostat linking plasma K+ levels to the activity of the Na-Cl cotransporter (NCC). This occurs through basolateral membrane potential sensing by inwardly rectifying K+ channels (Kir4.1/5.1); decrease in intracellular Cl-; activation of WNK4 and interaction and phosphorylation of STE20/SPS1-related proline/alanine-rich kinase (SPAK); binding of calcium-binding protein 39 (cab39) adaptor protein to SPAK, leading to its trafficking to the apical membrane; and SPAK binding, phosphorylation, and activation of NCC. As kidney-specific with-no-lysine kinase 1 (WNK1) isoform (KS-WNK1) is another participant in this pathway, we examined its function in NCC regulation. We eliminated KS-WNK1 specifically in the DCT and demonstrated increased expression of WNK4 and long WNK1 (L-WNK1) and increased phosphorylation of NCC. As in other KS-WNK1 models, the mice were not hyperkalemic. Although wild-type mice under low-dietary K+ conditions demonstrated increased NCC phosphorylation, the phosphorylation levels of the transporter, already high in KS-WNK1, did not change under the low-K+ diet. Thus, in the absence of KS-WNK1, the transporter lost its sensitivity to low plasma K+. We also show that under low K+ conditions, in the absence of KS-WNK1, there was no formation of WNK bodies. These bodies were observed in adjacent segments, not affected by the targeting of KS-WNK1. As our data are overall consistent with those of the global KS-WNK1 knockout, they indicate that the DCT is the predominant segment affecting the salt transport regulated by KS-WNK1.NEW & NOTEWORTHY In this paper, we show that KS-WNK1 is a critical component of the distal convoluted tubule (DCT) K+ switch pathway. Its deletion results in an inability of the DCT to sense changes in plasma potassium. Absence of KS-WNK1 leads to abnormally high levels of WNK4 and L-WNK1 in the DCT, resulting in increased Na-Cl phosphorylation and function. Our data are consistent with KS-WNK1 targeting WNK4 and L-WNK1 to degradation.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Andrew S Terker
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Rainelli B Koumangoye
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Lama Al-Qusairi
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Paul A Welling
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| |
Collapse
|
6
|
Omage K, McCormick JA. Cullin 3/with No Lysine [K] Kinase/Ste20/SPS-Related Proline Alanine Rich Kinase Signaling: Impact on NaCl Cotransporter Activity in BP Regulation. KIDNEY360 2024; 5:1386-1393. [PMID: 39120943 PMCID: PMC11441819 DOI: 10.34067/kid.0000000000000527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/11/2024]
Abstract
The sodium chloride cotransporter (NCC) fine-tunes Na + balance and indirectly affects the homeostasis of other ions including K + , Mg 2+ , and Ca 2+ . Owing to its effects on Na + balance, BP is significantly affected by alterations in NCC activity. Several factors have been reported to influence the expression and activity of NCC. One critical factor is NCC phosphorylation/dephosphorylation that occurs at key serine-threonine amino acid residues of the protein. Phosphorylation, which results in increased NCC activity, is mediated by the with no lysine [K] (WNK)-SPS-related proline alanine rich kinase (SPAK)/OSR1 kinases. NCC activation stimulates reabsorption of Na + , increasing extracellular fluid volume and hence BP. On the other hand, proteasomal degradation of WNK kinases after ubiquitination by the Cullin 3-Kelch-like 3 E3 ubiquitin ligase complex and dephosphorylation pathways oppose WNK-SPAK/OSR1-mediated NCC activation. Components of the Cullin 3/Kelch-like 3-WNK-SPAK/OSR1 regulatory pathway may be targets for novel antihypertensive drugs. In this review, we outline the impact of these regulators on the activity of NCC and the consequent effect on BP.
Collapse
Affiliation(s)
- Kingsley Omage
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | | |
Collapse
|
7
|
Rioux AV, Nsimba-Batomene TR, Slimani S, Bergeron NAD, Gravel MAM, Schreiber SV, Fiola MJ, Haydock L, Garneau AP, Isenring P. Navigating the multifaceted intricacies of the Na +-Cl - cotransporter, a highly regulated key effector in the control of hydromineral homeostasis. Physiol Rev 2024; 104:1147-1204. [PMID: 38329422 PMCID: PMC11381001 DOI: 10.1152/physrev.00027.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/01/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
The Na+-Cl- cotransporter (NCC; SLC12A3) is a highly regulated integral membrane protein that is known to exist as three splice variants in primates. Its primary role in the kidney is to mediate the cosymport of Na+ and Cl- across the apical membrane of the distal convoluted tubule. Through this role and the involvement of other ion transport systems, NCC allows the systemic circulation to reclaim a fraction of the ultrafiltered Na+, K+, Cl-, and Mg+ loads in exchange for Ca2+ and [Formula: see text]. The physiological relevance of the Na+-Cl- cotransport mechanism in humans is illustrated by several abnormalities that result from NCC inactivation through the administration of thiazides or in the setting of hereditary disorders. The purpose of the present review is to discuss the molecular mechanisms and overall roles of Na+-Cl- cotransport as the main topics of interest. On reading the narrative proposed, one will realize that the knowledge gained in regard to these themes will continue to progress unrelentingly no matter how refined it has now become.
Collapse
Affiliation(s)
- A V Rioux
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - T R Nsimba-Batomene
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - N A D Bergeron
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M A M Gravel
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S V Schreiber
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M J Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - L Haydock
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - A P Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - P Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
8
|
Zambrano-Carrasco J, Zou J, Wang W, Sun X, Li J, Su H. Emerging Roles of Cullin-RING Ubiquitin Ligases in Cardiac Development. Cells 2024; 13:235. [PMID: 38334627 PMCID: PMC10854628 DOI: 10.3390/cells13030235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Heart development is a spatiotemporally regulated process that extends from the embryonic phase to postnatal stages. Disruption of this highly orchestrated process can lead to congenital heart disease or predispose the heart to cardiomyopathy or heart failure. Consequently, gaining an in-depth understanding of the molecular mechanisms governing cardiac development holds considerable promise for the development of innovative therapies for various cardiac ailments. While significant progress in uncovering novel transcriptional and epigenetic regulators of heart development has been made, the exploration of post-translational mechanisms that influence this process has lagged. Culling-RING E3 ubiquitin ligases (CRLs), the largest family of ubiquitin ligases, control the ubiquitination and degradation of ~20% of intracellular proteins. Emerging evidence has uncovered the critical roles of CRLs in the regulation of a wide range of cellular, physiological, and pathological processes. In this review, we summarize current findings on the versatile regulation of cardiac morphogenesis and maturation by CRLs and present future perspectives to advance our comprehensive understanding of how CRLs govern cardiac developmental processes.
Collapse
Affiliation(s)
- Josue Zambrano-Carrasco
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Jianqiu Zou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Wenjuan Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA;
| | - Jie Li
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (J.Z.-C.); (J.Z.)
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Sigmund CD. The 2023 Walter B. Cannon Award Lecture: Mechanisms Regulating Vascular Function and Blood Pressure by the PPARγ-RhoBTB1-CUL3 Pathway. FUNCTION 2024; 5:zqad071. [PMID: 38196837 PMCID: PMC10775765 DOI: 10.1093/function/zqad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 01/11/2024] Open
Abstract
Human genetic and clinical trial data suggest that peroxisome proliferator activated receptor γ (PPARγ), a nuclear receptor transcription factor plays an important role in the regulation of arterial blood pressure. The examination of a series of novel animal models, coupled with transcriptomic and proteomic analysis, has revealed that PPARγ and its target genes employ diverse pathways to regulate vascular function and blood pressure. In endothelium, PPARγ target genes promote an antioxidant state, stimulating both nitric oxide (NO) synthesis and bioavailability, essential components of endothelial-smooth muscle communication. In vascular smooth muscle, PPARγ induces the expression of a number of genes that promote an antiinflammatory state and tightly control the level of cGMP, thus promoting responsiveness to endothelial-derived NO. One of the PPARγ targets in smooth muscle, Rho related BTB domain containing 1 (RhoBTB1) acts as a substrate adaptor for proteins to be ubiquitinated by the E3 ubiquitin ligase Cullin-3 and targeted for proteasomal degradation. One of these proteins, phosphodiesterase 5 (PDE5) is a target of the Cullin-3/RhoBTB1 pathway. Phosphodiesterase 5 degrades cGMP to GMP and thus regulates the smooth muscle response to NO. Moreover, expression of RhoBTB1 under condition of RhoBTB1 deficiency reverses established arterial stiffness. In conclusion, the coordinated action of PPARγ in endothelium and smooth muscle is needed to maintain NO bioavailability and activity, is an essential regulator of vasodilator/vasoconstrictor balance, and regulates blood vessel structure and stiffness.
Collapse
Affiliation(s)
- Curt D Sigmund
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
10
|
Maeoka Y, Nguyen LT, Sharma A, Cornelius RJ, Su XT, Gutierrez MR, Carbajal-Contreras H, Castañeda-Bueno M, Gamba G, McCormick JA. Dysregulation of the WNK4-SPAK/OSR1 pathway has a minor effect on baseline NKCC2 phosphorylation. Am J Physiol Renal Physiol 2024; 326:F39-F56. [PMID: 37881876 PMCID: PMC11905798 DOI: 10.1152/ajprenal.00100.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
The with-no-lysine kinase 4 (WNK4)-sterile 20/SPS-1-related proline/alanine-rich kinase (SPAK)/oxidative stress-responsive kinase 1 (OSR1) pathway mediates activating phosphorylation of the furosemide-sensitive Na+-K+-2Cl- cotransporter (NKCC2) and the thiazide-sensitive NaCl cotransporter (NCC). The commonly used pT96/pT101-pNKCC2 antibody cross-reacts with pT53-NCC in mice on the C57BL/6 background due to a five amino acid deletion. We generated a new C57BL/6-specific pNKCC2 antibody (anti-pT96-NKCC2) and tested the hypothesis that the WNK4-SPAK/OSR1 pathway strongly regulates the phosphorylation of NCC but not NKCC2. In C57BL/6 mice, anti-pT96-NKCC2 detected pNKCC2 and did not cross-react with NCC. Abundances of pT96-NKCC2 and pT53-NCC were evaluated in Wnk4-/-, Osr1-/-, Spak-/-, and Osr1-/-/Spak-/- mice and in several models of the disease familial hyperkalemic hypertension (FHHt) in which the CUL3-KLHL3 ubiquitin ligase complex that promotes WNK4 degradation is dysregulated (Cul3+/-/Δ9, Klhl3-/-, and Klhl3R528H/R528H). All mice were on the C57BL/6 background. In Wnk4-/- mice, pT53-NCC was almost absent but pT96-NKCC2 was only slightly lower. pT53-NCC was almost absent in Spak-/- and Osr1-/-/Spak-/- mice, but pT96-NKCC2 abundance did not differ from controls. pT96-NKCC2/total NKCC2 was slightly lower in Osr1-/- and Osr1-/-/Spak-/- mice. WNK4 expression colocalized not only with NCC but also with NKCC2 in Klhl3-/- mice, but pT96-NKCC2 abundance was unchanged. Consistent with this, furosemide-induced urinary Na+ excretion following thiazide treatment was similar between Klhl3-/- and controls. pT96-NKCC2 abundance was also unchanged in the other FHHt mouse models. Our data show that disruption of the WNK4-SPAK/OSR1 pathway only mildly affects NKCC2 phosphorylation, suggesting a role for other kinases in NKCC2 activation. In FHHt models NKCC2 phosphorylation is unchanged despite higher WNK4 abundance, explaining the thiazide sensitivity of FHHt.NEW & NOTEWORTHY The renal cation cotransporters NCC and NKCC2 are activated following phosphorylation mediated by the WNK4-SPAK/OSR1 pathway. While disruption of this pathway strongly affects NCC activity, effects on NKCC2 activity are unclear since the commonly used phospho-NKCC2 antibody was recently reported to cross-react with phospho-NCC in mice on the C57BL/6 background. Using a new phospho-NKCC2 antibody specific for C57BL/6, we show that inhibition or activation of the WNK4-SPAK/OSR1 pathway in mice only mildly affects NKCC2 phosphorylation.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Luan T Nguyen
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Marissa R Gutierrez
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
11
|
Sharma P, Chatrathi HE. Insights into the diverse mechanisms and effects of variant CUL3-induced familial hyperkalemic hypertension. Cell Commun Signal 2023; 21:286. [PMID: 37845702 PMCID: PMC10577937 DOI: 10.1186/s12964-023-01269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/12/2023] [Indexed: 10/18/2023] Open
Abstract
Familial hyperkalemic hypertension (FHHt), also known as Pseudohypoaldosteronism type II (PHAII) or Gordon syndrome is a rare Mendelian disease classically characterized by hyperkalemia, hyperchloremic metabolic acidosis, and high systolic blood pressure. The most severe form of the disease is caused by autosomal dominant variants in CUL3 (Cullin 3), a critical subunit of the multimeric CUL3-RING ubiquitin ligase complex. The recent identification of a novel FHHt disease variant of CUL3 revealed intricacies within the underlying disease mechanism. When combined with studies on canonical CUL3 variant-induced FHHt, these findings further support CUL3's role in regulating renal electrolyte transport and maintaining systemic vascular tone. However, the pathophysiological effects of CUL3 variants are often accompanied by diverse systemic disturbances in addition to classical FHHt symptoms. Recent global proteomic analyses provide a rationale for these systemic disturbances, paving the way for future mechanistic studies to reveal how CUL3 variants dysregulate processes outside of the renovascular axis. Video Abstract.
Collapse
Affiliation(s)
- Prashant Sharma
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, USA.
| | - Harish E Chatrathi
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
12
|
Cornelius RJ, Maeoka Y, McCormick JA. Renal effects of cullin 3 mutations causing familial hyperkalemic hypertension. Curr Opin Nephrol Hypertens 2023; 32:335-343. [PMID: 37070483 PMCID: PMC10330058 DOI: 10.1097/mnh.0000000000000891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
PURPOSE OF REVIEW Mutations in the E3 ubiquitin ligase scaffold cullin 3 (CUL3) cause the disease familial hyperkalemic hypertension (FHHt) by hyperactivating the NaCl cotransporter (NCC). The effects of these mutations are complex and still being unraveled. This review discusses recent findings revealing the molecular mechanisms underlying the effects of CUL3 mutations in the kidney. RECENT FINDINGS The naturally occurring mutations that cause deletion of exon 9 (CUL3-Δ9) from CUL3 generate an abnormal CUL3 protein. CUL3-Δ9 displays increased interaction with multiple ubiquitin ligase substrate adaptors. However, in-vivo data show that the major mechanism for disease pathogenesis is that CUL3-Δ9 promotes degradation of itself and KLHL3, the specific substrate adaptor for an NCC-activating kinase. CUL3-Δ9 displays dysregulation via impaired binding to the CSN and CAND1, which cause hyperneddylation and compromised adaptor exchange, respectively. A recently discovered CUL3 mutant (CUL3-Δ474-477) displays many similarities to CUL3-Δ9 mutations but some key differences that likely account for the milder FHHt phenotype it elicits. Furthermore, recent work suggests that CUL3 mutations could have unidentified complications in patients and/or a predisposition to renal injury. SUMMARY This review summarizes recent studies highlighting advances in our understanding of the renal mechanisms by which CUL3 mutations modulate blood pressure in FHHt.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | | | | |
Collapse
|
13
|
Maeoka Y, Cornelius RJ, McCormick JA. Cullin 3 and Blood Pressure Regulation: Insights From Familial Hyperkalemic Hypertension. Hypertension 2023; 80:912-923. [PMID: 36861484 PMCID: PMC10133098 DOI: 10.1161/hypertensionaha.123.20525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The study of rare monogenic forms of hypertension has led to the elucidation of important physiological pathways controlling blood pressure. Mutations in several genes cause familial hyperkalemic hypertension (also known as Gordon syndrome or pseudohypoaldosteronism type II). The most severe form of familial hyperkalemic hypertension is caused by mutations in CUL3, encoding CUL3 (Cullin 3)-a scaffold protein in an E3 ubiquitin ligase complex that tags substrates for proteasomal degradation. In the kidney, CUL3 mutations cause accumulation of the substrate WNK (with-no-lysine [K]) kinase and ultimately hyperactivation of the renal NaCl cotransporter-the target of the first-line antihypertensive thiazide diuretics. The precise mechanisms by which mutant CUL3 causes WNK kinase accumulation have been unclear, but several functional defects are likely to contribute. The hypertension seen in familial hyperkalemic hypertension also results from effects exerted by mutant CUL3 on several pathways in vascular smooth muscle and endothelium that modulate vascular tone. This review summarizes the mechanisms by which wild type and mutant CUL3 modulate blood pressure through effects on the kidney and vasculature, potential effects in the central nervous system and heart, and future directions for investigation.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR
| |
Collapse
|
14
|
Maeoka Y, Cornelius RJ, Ferdaus MZ, Sharma A, Nguyen LT, McCormick JA. Cullin 3 mutant causing familial hyperkalemic hypertension lacks normal activity in the kidney. Am J Physiol Renal Physiol 2022; 323:F564-F576. [PMID: 36007890 PMCID: PMC9602935 DOI: 10.1152/ajprenal.00153.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 08/15/2022] [Indexed: 01/07/2023] Open
Abstract
Mutations in the ubiquitin ligase scaffold protein cullin 3 (CUL3) cause the disease familial hyperkalemic hypertension (FHHt). We recently reported that in the kidney, aberrant mutant CUL3 (CUL3-Δ9) activity lowers the abundance of CUL3-Δ9 and Kelch-like 3, the CUL3 substrate adaptor for with-no-lysine kinase 4 (WNK4) and that this is mechanistically important. However, whether CUL3-Δ9 exerts additional effects on other targets that may alter renal function is unclear. Here, we sought to determine 1) whether CUL3-Δ9 expression can rescue the phenotype of renal tubule-specific Cul3 knockout mice, and 2) whether CUL3-Δ9 expression affects other CUL3 substrates. Using an inducible renal tubule-specific system, we studied two CUL3-Δ9-expressing mouse models: Cul3 knockout (Cul3-/-/Δ9) and Cul3 heterozygous background (Cul3+/-/Δ9, FHHt model). The effects of CUL3-Δ9 in these mice were compared with Cul3-/- and Cul3+/- mice. Similar to Cul3-/- mice, Cul3-/-/Δ9 mice displayed polyuria with loss of aquaporin 2 and collecting duct injury; proximal tubule injury also occurred. CUL3-Δ9 did not promote degradation of two CUL3 targets that accumulate in the Cul3-/- kidney: high-molecular-weight (HMW) cyclin E and NAD(P)H:quinone oxidoreductase 1 (NQO1) [a surrogate for the CUL3-Kelch-like ECH-associated protein 1 (KEAP1) substrate nuclear factor erythroid-2-related factor 2]. Since CUL3-Δ9 expression cannot rescue the Cul3-/- phenotype, our data suggest that CUL3-Δ9 cannot normally function in ubiquitin ligase complexes. In Cul3+/-/Δ9 mice, KEAP1 abundance did not differ but NQO1 abundance was higher, suggesting adaptor sequestration by CUL3-Δ9 in vivo. Together, our results provide evidence that in the kidney, CUL3-Δ9 completely lacks normal activity and can trap CUL3 substrate adaptors in inactive complexes.NEW & NOTEWORTHY CUL3 mutation (CUL3-Δ9) causes familial hyperkalemic hypertension (FHHt) by reducing adaptor KLHL3, impairing substrate WNK4 degradation. Whether CUL3-Δ9 affects other targets in kidneys remains unclear. We found that CUL3-Δ9 cannot degrade two CUL3 targets, cyclin E and nuclear factor erythroid-2-related factor 2 (NRF2; using a surrogate marker NQO1), or rescue injury or polyuria caused by Cul3 disruption. In an FHHt model, CUL3-Δ9 impaired NRF2 degradation without reduction of its adaptor KEAP1. Our data provide additional insights into CUL3-Δ9 function in the kidney.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Mohammed Zubaerul Ferdaus
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Luan T Nguyen
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
15
|
Cullin 3 Exon 9 Deletion in Familial Hyperkalemic Hypertension Impairs Cullin3-Ring-E3 Ligase (CRL3) Dynamic Regulation and Cycling. Int J Mol Sci 2022; 23:ijms23095151. [PMID: 35563538 PMCID: PMC9105235 DOI: 10.3390/ijms23095151] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 02/06/2023] Open
Abstract
Cullin 3 (CUL3) is the scaffold of Cullin3 Ring E3-ligases (CRL3s), which use various BTB-adaptor proteins to ubiquitinate numerous substrates targeting their proteasomal degradation. CUL3 mutations, responsible for a severe form of familial hyperkalemia and hypertension (FHHt), all result in a deletion of exon 9 (amino-acids 403-459) (CUL3-∆9). Surprisingly, while CUL3-∆9 is hyperneddylated, a post-translational modification that typically activates CRL complexes, it is unable to ubiquitinate its substrates. In order to understand the mechanisms behind this loss-of function, we performed comparative label-free quantitative analyses of CUL3 and CUL3-∆9 interactome by mass spectrometry. It was observed that CUL3-∆9 interactions with COP9 and CAND1, both involved in CRL3 complexes’ dynamic assembly, were disrupted. These defects result in a reduction in the dynamic cycling of the CRL3 complexes, making the CRL3-∆9 complex an inactive BTB-adaptor trap, as demonstrated by SILAC experiments. Collectively, the data indicated that the hyperneddylated CUL3-∆9 protein is inactive as a consequence of several structural changes disrupting its dynamic interactions with key regulatory partners.
Collapse
|
16
|
Porter AW, Nguyen DN, Clayton DR, Ruiz WG, Mutchler SM, Ray EC, Marciszyn AL, Nkashama LJ, Subramanya AR, Gingras S, Kleyman TR, Apodaca G, Hendershot LM, Brodsky JL, Buck TM. The molecular chaperone GRP170 protects against ER stress and acute kidney injury in mice. JCI Insight 2022; 7:e151869. [PMID: 35104250 PMCID: PMC8983141 DOI: 10.1172/jci.insight.151869] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/28/2022] [Indexed: 01/26/2023] Open
Abstract
Molecular chaperones are responsible for maintaining cellular homeostasis, and one such chaperone, GRP170, is an endoplasmic reticulum (ER) resident that oversees both protein biogenesis and quality control. We previously discovered that GRP170 regulates the degradation and assembly of the epithelial sodium channel (ENaC), which reabsorbs sodium in the distal nephron and thereby regulates salt-water homeostasis and blood pressure. To define the role of GRP170 - and, more generally, molecular chaperones in kidney physiology - we developed an inducible, nephron-specific GRP170-KO mouse. Here, we show that GRP170 deficiency causes a dramatic phenotype: profound hypovolemia, hyperaldosteronemia, and dysregulation of ion homeostasis, all of which are associated with the loss of ENaC. Additionally, the GRP170-KO mouse exhibits hallmarks of acute kidney injury (AKI). We further demonstrate that the unfolded protein response (UPR) is activated in the GRP170-deficient mouse. Notably, the UPR is also activated in AKI when originating from various other etiologies, including ischemia, sepsis, glomerulonephritis, nephrotic syndrome, and transplant rejection. Our work establishes the central role of GRP170 in kidney homeostasis and directly links molecular chaperone function to kidney injury.
Collapse
Affiliation(s)
- Aidan W. Porter
- Department of Biological Sciences
- Department of Pediatrics, Nephrology Division
| | | | | | - Wily G. Ruiz
- Department of Medicine, Renal-Electrolyte Division
| | | | - Evan C. Ray
- Department of Medicine, Renal-Electrolyte Division
| | | | | | | | | | - Thomas R. Kleyman
- Department of Medicine, Renal-Electrolyte Division
- Department of Cell Biology, and
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
17
|
Maeoka Y, Ferdaus MZ, Cornelius RJ, Sharma A, Su XT, Miller LN, Robertson JA, Gurley SB, Yang CL, Ellison DH, McCormick JA. Combined Kelch-like 3 and Cullin 3 Degradation is a Central Mechanism in Familial Hyperkalemic Hypertension in Mice. J Am Soc Nephrol 2022; 33:584-600. [PMID: 35064051 PMCID: PMC8975056 DOI: 10.1681/asn.2021081099] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/10/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) gene cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) increases abundance of With-No-Lysine (K) Kinase 4 (WNK4), inappropriately activating sterile 20/SPS-1-related proline/alanine-rich kinase (SPAK), which then phosphorylates and hyperactivates the Na+Cl- cotransporter (NCC). The precise mechanism by which CUL3-Δ9 causes FHHt is unclear. We tested the hypothesis that reduced abundance of CUL3 and of Kelch-like 3 (KLHL3), the CUL3 substrate adaptor for WNK4, is mechanistically important. Because JAB1, an enzyme that inhibits CUL3 activity by removing the ubiquitin-like protein NEDD8, cannot interact with CUL3-Δ9, we also determined whether Jab1 disruption mimicked the effects of CUL3-Δ9 expression. METHODS We used an inducible renal tubule-specific system to generate several mouse models expressing CUL3-Δ9, mice heterozygous for both CUL3 and KLHL3 (Cul3+/-/Klhl3+/- ), and mice with short-term Jab1 disruption (to avoid renal injury associated with long-term disruption). RESULTS Renal KLHL3 was higher in Cul3-/- mice, but lower in Cul3-/-/Δ9 mice and in the Cul3+/-/Δ9 FHHt model, suggesting KLHL3 is a target for both WT and mutant CUL3. Cul3+/-/Klhl3+/- mice displayed increased WNK4-SPAK activation and phospho-NCC abundance and an FHHt-like phenotype with increased plasma [K+] and salt-sensitive blood pressure. Short-term Jab1 disruption in mice lowered the abundance of CUL3 and KLHL3 and increased the abundance of WNK4 and phospho-NCC. CONCLUSIONS Jab1-/- mice and Cul3+/-/Klhl3+/- mice recapitulated the effects of CUL3-Δ9 expression on WNK4-SPAK-NCC. Our data suggest degradation of both KLHL3 and CUL3 plays a central mechanistic role in CUL3-Δ9-mediated FHHt.
Collapse
Affiliation(s)
- Yujiro Maeoka
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Mohammed Z. Ferdaus
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Ryan J. Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Lauren N. Miller
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joshua A. Robertson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Susan B. Gurley
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
- Veterans Affairs Portland Healthcare System, Portland, Oregon
| | - James A. McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
18
|
Chatrathi HE, Collins JC, Wolfe LA, Markello TC, Adams DR, Gahl WA, Werner A, Sharma P. Novel CUL3 Variant Causing Familial Hyperkalemic Hypertension Impairs Regulation and Function of Ubiquitin Ligase Activity. Hypertension 2022; 79:60-75. [PMID: 34878901 PMCID: PMC8667186 DOI: 10.1161/hypertensionaha.121.17624] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Familial hyperkalemic hypertension is caused by pathogenic variants in genes of the CUL3 (cullin-3)-KLHL3 (kelch-like-family-member-3)-WNK (with no-lysine [K] kinase) pathway, manifesting clinically as hyperkalemia, metabolic acidosis, and high systolic blood pressure. The ubiquitin E3 ligase CUL3-KLHL3 targets WNK kinases for degradation to limit activation of the thiazide-sensitive NCC (Na-Cl cotransporter). All known variants in CUL3 lead to exon 9 skipping (CUL3Δ9) and typically result in severe familial hyperkalemic hypertension and growth disturbances in patients. Whether other variants in CUL3 cause familial hyperkalemic hypertension is unknown. Here, we identify a novel de novo heterozygous CUL3 variant (CUL3Δ474-477) in a pediatric familial hyperkalemic hypertension patient with multiple congenital anomalies and reveal molecular mechanisms by which CUL3Δ474-477 leads to dysregulation of the CUL3-KLHL3-WNK signaling axis. Using patient-derived urinary extracellular vesicles and dermal fibroblasts, in vitro assays, and cultured kidney cells, we demonstrate that CUL3Δ474-477 causes reduced total CUL3 levels due to increased autoubiquitination. The CUL3Δ474-477 that escapes autodegradation shows enhanced modification with NEDD8 (neural precursor cell expressed developmentally down-regulated protein 8) and increased formation of CUL3-KLHL3 complexes that are impaired in ubiquitinating WNK4. Proteomic analysis of CUL3 complexes revealed that, in addition to increased KLHL3 binding, the CUL3Δ474-477 variant also exhibits increased interactions with other BTB (Bric-a-brac, Tramtrack, and Broad complex) substrate adaptors, providing a rationale for the patient's diverse phenotypes. We conclude that the pathophysiological effects of CUL3Δ474-477 are caused by reduced CUL3 levels and formation of catalytically impaired CUL3 ligase complexes.
Collapse
Affiliation(s)
- Harish E. Chatrathi
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA,Share the first authorship position
| | - Jason C. Collins
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA,Share the first authorship position
| | - Lynne A. Wolfe
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Thomas C. Markello
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA,Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland Bethesda, Maryland 20892, USA
| | - David R. Adams
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA,Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland Bethesda, Maryland 20892, USA
| | - William A. Gahl
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA,Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland Bethesda, Maryland 20892, USA
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Prashant Sharma
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, Maryland 20892, USA,Correspondence: Prashant Sharma, NIH Undiagnosed Diseases Program, National Human Genome Research Institute, 5625 Fishers Lane, Rockville, MD.
| |
Collapse
|
19
|
Potassium Effects on NCC Are Attenuated during Inhibition of Cullin E3-Ubiquitin Ligases. Cells 2021; 11:cells11010095. [PMID: 35011657 PMCID: PMC8750104 DOI: 10.3390/cells11010095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 01/02/2023] Open
Abstract
The thiazide-sensitive sodium chloride cotransporter (NCC) plays a vital role in maintaining sodium (Na+) and potassium (K+) homeostasis. NCC activity is modulated by with-no-lysine kinases 1 and 4 (WNK1 and WNK4), the abundance of which is controlled by the RING-type E3 ligase Cullin 3 (Cul3) and its substrate adapter Kelch-like protein 3. Dietary K+ intake has an inverse correlation with NCC activity, but the mechanism underlying this phenomenon remains to be fully elucidated. Here, we investigated the involvement of other members of the cullin family in mediating K+ effects on NCC phosphorylation (active form) and abundance. In kidneys from mice fed diets varying in K+ content, there were negative correlations between NCC (phosphorylated and total) and active (neddylated) forms of cullins (Cul1, 3, 4, and 5). High dietary K+ effects on phosphorylated NCC were attenuated in Cul3 mutant mice (CUL3-Het/Δ9). Short-term (30 min) and long-term (24 h) alterations in the extracellular K+ concentration did not affect cullin neddylation levels in ex vivo renal tubules. In the short term, the ability of high extracellular K+ to decrease NCC phosphorylation was preserved in the presence of MLN4924 (pan-cullin inhibitor), but the response to low extracellular K+ was absent. In the long term, MLN4924 attenuated the effects of high extracellular K+ on NCC phosphorylation, and responses to low extracellular K+ were absent. Our data suggest that in addition to Cul3, other cullins are involved in mediating the effects of K+ on NCC phosphorylation and abundance.
Collapse
|
20
|
Marcoux AA, Tremblay LE, Slimani S, Fiola MJ, Mac-Way F, Haydock L, Garneau AP, Isenring P. Anatomophysiology of the Henle's Loop: Emphasis on the Thick Ascending Limb. Compr Physiol 2021; 12:3119-3139. [PMID: 34964111 DOI: 10.1002/cphy.c210021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The loop of Henle plays a variety of important physiological roles through the concerted actions of ion transport systems in both its apical and basolateral membranes. It is involved most notably in extracellular fluid volume and blood pressure regulation as well as Ca2+ , Mg2+ , and acid-base homeostasis because of its ability to reclaim a large fraction of the ultrafiltered solute load. This nephron segment is also involved in urinary concentration by energizing several of the steps that are required to generate a gradient of increasing osmolality from cortex to medulla. Another important role of the loop of Henle is to sustain a process known as tubuloglomerular feedback through the presence of specialized renal tubular cells that lie next to the juxtaglomerular arterioles. This article aims at describing these physiological roles and at discussing a number of the molecular mechanisms involved. It will also report on novel findings and uncertainties regarding the realization of certain processes and on the pathophysiological consequences of perturbed salt handling by the thick ascending limb of the loop of Henle. Since its discovery 150 years ago, the loop of Henle has remained in the spotlight and is now generating further interest because of its role in the renal-sparing effect of SGLT2 inhibitors. © 2022 American Physiological Society. Compr Physiol 12:1-21, 2022.
Collapse
Affiliation(s)
- Andrée-Anne Marcoux
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Laurence E Tremblay
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Samira Slimani
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Marie-Jeanne Fiola
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Fabrice Mac-Way
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Ludwig Haydock
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| | - Alexandre P Garneau
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada.,Cardiometabolic Axis, School of Kinesiology and Physical Activity Sciences, University of Montréal, Montréal, QC, Canada
| | - Paul Isenring
- Nephrology Research Group, Department of Medicine, Laval University, Québec, QC, Canada
| |
Collapse
|
21
|
Castañeda-Bueno M, Ellison DH, Gamba G. Molecular mechanisms for the modulation of blood pressure and potassium homeostasis by the distal convoluted tubule. EMBO Mol Med 2021; 14:e14273. [PMID: 34927382 PMCID: PMC8819348 DOI: 10.15252/emmm.202114273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Epidemiological and clinical observations have shown that potassium ingestion is inversely correlated with arterial hypertension prevalence and cardiovascular mortality. The higher the dietary potassium, the lower the blood pressure and mortality. This phenomenon is explained, at least in part, by the interaction between salt reabsorption in the distal convoluted tubule (DCT) and potassium secretion in the connecting tubule/collecting duct of the mammalian nephron: In order to achieve adequate K+ secretion levels under certain conditions, salt reabsorption in the DCT must be reduced. Because salt handling by the kidney constitutes the basis for the long‐term regulation of blood pressure, losing salt prevents hypertension. Here, we discuss how the study of inherited diseases in which salt reabsorption in the DCT is affected has revealed the molecular players, including membrane transporters and channels, kinases, and ubiquitin ligases that form the potassium sensing mechanism of the DCT and the processes through which the consequent adjustments in salt reabsorption are achieved.
Collapse
Affiliation(s)
- María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, OR, USA.,Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, OR, USA.,VA Portland Health Care System, Portland, OR, USA
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| |
Collapse
|
22
|
Morandell J, Schwarz LA, Basilico B, Tasciyan S, Dimchev G, Nicolas A, Sommer C, Kreuzinger C, Dotter CP, Knaus LS, Dobler Z, Cacci E, Schur FKM, Danzl JG, Novarino G. Cul3 regulates cytoskeleton protein homeostasis and cell migration during a critical window of brain development. Nat Commun 2021; 12:3058. [PMID: 34031387 PMCID: PMC8144225 DOI: 10.1038/s41467-021-23123-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/14/2021] [Indexed: 01/03/2023] Open
Abstract
De novo loss of function mutations in the ubiquitin ligase-encoding gene Cullin3 (CUL3) lead to autism spectrum disorder (ASD). In mouse, constitutive Cul3 haploinsufficiency leads to motor coordination deficits as well as ASD-relevant social and cognitive impairments. However, induction of Cul3 haploinsufficiency later in life does not lead to ASD-relevant behaviors, pointing to an important role of Cul3 during a critical developmental window. Here we show that Cul3 is essential to regulate neuronal migration and, therefore, constitutive Cul3 heterozygous mutant mice display cortical lamination abnormalities. At the molecular level, we found that Cul3 controls neuronal migration by tightly regulating the amount of Plastin3 (Pls3), a previously unrecognized player of neural migration. Furthermore, we found that Pls3 cell-autonomously regulates cell migration by regulating actin cytoskeleton organization, and its levels are inversely proportional to neural migration speed. Finally, we provide evidence that cellular phenotypes associated with autism-linked gene haploinsufficiency can be rescued by transcriptional activation of the intact allele in vitro, offering a proof of concept for a potential therapeutic approach for ASDs. De novo loss of function mutations in the ubiquitin ligase-encoding gene Cullin3 (CUL3) lead to autism spectrum disorder (ASD). Here, the authors show that Cul3 is essential to regulate neuronal migration by tightly regulating Plastin3 (Pls3). Pls3 cell-autonomously regulates cell migration by regulating the actin cytoskeleton organization.
Collapse
Affiliation(s)
- Jasmin Morandell
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Lena A Schwarz
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | | | - Saren Tasciyan
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Georgi Dimchev
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Armel Nicolas
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | | | - Christoph P Dotter
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Lisa S Knaus
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Zoe Dobler
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Emanuele Cacci
- Department of Biology and Biotechnology "Charles Darwin", Sapienza, University of Rome, Rome, Italy
| | - Florian K M Schur
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Johann G Danzl
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria.
| |
Collapse
|
23
|
Zhang Y, Jiang G, Zhang C. Downregulation of Cullin 3 Ligase Signaling Pathways Contributes to Hypertension in Preeclampsia. Front Cardiovasc Med 2021; 8:654254. [PMID: 33928137 PMCID: PMC8076533 DOI: 10.3389/fcvm.2021.654254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/12/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Preeclampsia (PE) is a leading cause of maternal and perinatal morbidity and mortality; however, its etiology and pathophysiology remain obscure. PE is initiated by inadequate spiral artery remodeling and subsequent placental ischemia/hypoxia, which stimulates release of bioactive factors into maternal circulation, leading to hypertension and renal damage. Methods and Results: Abundance of key components of cullin 3-ring ubiquitin ligase (CRL3), including cullin 3 (CUL3) and its neddylated modification, and adaptors including Kelch-like 2 (KLHL2) and Rho-related BTB domain containing protein 1 was all decreased in spiral arteries and placentas of PE patients. Similar changes were found in aortic tissues and renal distal tubules of pregnant mice treated with Nω-nitro-l-arginine methyl ester hydrochloride. The downregulation of CRL3 function led to accumulation of with-no-lysine kinases, phosphodiesterase 5, and RhoA in vessels and renal distal tubules, which promoted vasoconstriction and Na-Cl cotransporter activation in the distal convoluted tubule (DCT), as well as vascular and DCT structure remodeling. Proton pump inhibitor esomeprazole partially restored CRL3 function. In vitro studies have shown that increased abundance of JAB1, a component of the COP9 signalosome, inhibited CUL3 neddylation and promoted the expression of hypoxia-inducible factor 1α, which downregulated peroxisome proliferator-activated receptor γ and further promoted CUL3 inactivation. KLHL3/2 was degraded by increased autophagy. Conclusion: These findings support that the downregulation of CRL3 function disrupts the balance of vasoconstriction and vasodilation and aggravates excess reabsorption of sodium in PE.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gengru Jiang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chong Zhang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Fang S, Livergood MC, Nakagawa P, Wu J, Sigmund CD. Role of the Peroxisome Proliferator Activated Receptors in Hypertension. Circ Res 2021; 128:1021-1039. [PMID: 33793338 DOI: 10.1161/circresaha.120.318062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors represent a large family of ligand-activated transcription factors which sense the physiological environment and make long-term adaptations by mediating changes in gene expression. In this review, we will first discuss the fundamental mechanisms by which nuclear receptors mediate their transcriptional responses. We will focus on the PPAR (peroxisome proliferator-activated receptor) family of adopted orphan receptors paying special attention to PPARγ, the isoform with the most compelling evidence as an important regulator of arterial blood pressure. We will review genetic data showing that rare mutations in PPARγ cause severe hypertension and clinical trial data which show that PPARγ activators have beneficial effects on blood pressure. We will detail the tissue- and cell-specific molecular mechanisms by which PPARs in the brain, kidney, vasculature, and immune system modulate blood pressure and related phenotypes, such as endothelial function. Finally, we will discuss the role of placental PPARs in preeclampsia, a life threatening form of hypertension during pregnancy. We will close with a viewpoint on future research directions and implications for developing novel therapies.
Collapse
Affiliation(s)
- Shi Fang
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee.,Department of Neuroscience and Pharmacology, University of Iowa (S.F.)
| | - M Christine Livergood
- Department of Obstetrics and Gynecology (M.C.L.), Medical College of Wisconsin, Milwaukee
| | - Pablo Nakagawa
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| | - Jing Wu
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| | - Curt D Sigmund
- Department of Physiology, Cardiovascular Center (S.F., P.N., J.W., C.D.S.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
25
|
Murillo-de-Ozores AR, Rodríguez-Gama A, Carbajal-Contreras H, Gamba G, Castañeda-Bueno M. WNK4 kinase: from structure to physiology. Am J Physiol Renal Physiol 2021; 320:F378-F403. [PMID: 33491560 DOI: 10.1152/ajprenal.00634.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With no lysine kinase-4 (WNK4) belongs to a serine-threonine kinase family characterized by the atypical positioning of its catalytic lysine. Despite the fact that WNK4 has been found in many tissues, the majority of its study has revolved around its function in the kidney, specifically as a positive regulator of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule of the nephron. This is explained by the description of gain-of-function mutations in the gene encoding WNK4 that causes familial hyperkalemic hypertension. This disease is mainly driven by increased downstream activation of the Ste20/SPS1-related proline-alanine-rich kinase/oxidative stress responsive kinase-1-NCC pathway, which increases salt reabsorption in the distal convoluted tubule and indirectly impairs renal K+ secretion. Here, we review the large volume of information that has accumulated about different aspects of WNK4 function. We first review the knowledge on WNK4 structure and enumerate the functional domains and motifs that have been characterized. Then, we discuss WNK4 physiological functions based on the information obtained from in vitro studies and from a diverse set of genetically modified mouse models with altered WNK4 function. We then review in vitro and in vivo evidence on the different levels of regulation of WNK4. Finally, we go through the evidence that has suggested how different physiological conditions act through WNK4 to modulate NCC activity.
Collapse
Affiliation(s)
- Adrián Rafael Murillo-de-Ozores
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | | | - Héctor Carbajal-Contreras
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico.,Combined Studies Program in Medicine MD/PhD (PECEM), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico, Mexico
| |
Collapse
|
26
|
Meor Azlan NF, Zhang J. Role of the Cation-Chloride-Cotransporters in Cardiovascular Disease. Cells 2020; 9:2293. [PMID: 33066544 PMCID: PMC7602155 DOI: 10.3390/cells9102293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 02/05/2023] Open
Abstract
The SLC12 family of cation-chloride-cotransporters (CCCs) is comprised of potassium chloride cotransporters (KCCs), which mediate Cl- extrusion and sodium-potassium chloride cotransporters (N[K]CCs), which mediate Cl- loading. The CCCs play vital roles in cell volume regulation and ion homeostasis. The functions of CCCs influence a variety of physiological processes, many of which overlap with the pathophysiology of cardiovascular disease. Although not all of the cotransporters have been linked to Mendelian genetic disorders, recent studies have provided new insights into their functional role in vascular and renal cells in addition to their contribution to cardiovascular diseases. Particularly, an imbalance in potassium levels promotes the pathogenesis of atherosclerosis and disturbances in sodium homeostasis are one of the causes of hypertension. Recent findings suggest hypothalamic signaling as a key signaling pathway in the pathophysiology of hypertension. In this review, we summarize and discuss the role of CCCs in cardiovascular disease with particular emphasis on knowledge gained in recent years on NKCCs and KCCs.
Collapse
Affiliation(s)
- Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, Hatherly Laboratories, University of Exeter, Exeter EX4 4PS, UK;
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The goal of this review is to evaluate recent advances in understanding the pivotal roles of Cullin-3 (CUL3) in blood pressure regulation with a focus on its actions in the kidney and blood vessels. RECENT FINDINGS Cul3-based ubiquitin ligase regulates renal electrolyte transport, vascular tone, and redox homeostasis by facilitating the normal turnover of (1) with-no-lysine kinases in the distal nephron, (2) RhoA and phosphodiesterase 5 in the vascular smooth muscle, and (3) nuclear factor E2-related factor 2 in antioxidant responses. CUL3 mutations identified in familial hyperkalemic hypertension (FHHt) yield a mutant protein lacking exon 9 (CUL3∆9) which displays dual gain and loss of function. CUL3∆9 acts in a dominant manner to impair CUL3-mediated substrate ubiquitylation and degradation. The consequent accumulation of substrates and overactivation of downstream signaling cause FHHt through increased sodium reabsorption, enhanced vasoconstriction, and decreased vasodilation. CUL3 ubiquitin ligase maintains normal cardiovascular and renal physiology through posttranslational modification of key substrates which regulate blood pressure. Interference with CUL3 disturbs these key downstream pathways. Further understanding the spatial and temporal specificity of how CUL3 functions in these pathways is necessary to identify novel therapeutic targets for hypertension.
Collapse
|
28
|
Cornelius RJ, Sharma A, Su XT, Guo JJ, McMahon JA, Ellison DH, McMahon AP, McCormick JA. A novel distal convoluted tubule-specific Cre-recombinase driven by the NaCl cotransporter gene. Am J Physiol Renal Physiol 2020; 319:F423-F435. [PMID: 32657158 DOI: 10.1152/ajprenal.00101.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cre-lox technology has revolutionized research in renal physiology by allowing site-specific genetic recombination in individual nephron segments. The distal convoluted tubule (DCT), consisting of distinct early (DCT1) and late (DCT2) segments, plays a central role in Na+ and K+ homeostasis. The only established Cre line targeting the DCT is Pvalb-Cre, which is limited by noninducibility, activity along DCT1 only, and activity in neurons. Here, we report the characterization of the first Cre line specific to the entire DCT. CRISPR/Cas9 targeting was used to introduce a tamoxifen-inducible IRES-Cre-ERT2 cassette downstream of the coding region of the Slc12a3 gene encoding the NaCl cotransporter (NCC). The resulting Slc12a3-Cre-ERT2 mice were crossed with R26R-YFP reporter mice, which revealed minimal leakiness with 6.3% of NCC-positive cells expressing yellow fluorescent protein (YFP) in the absence of tamoxifen. After tamoxifen injection, YFP expression was observed in 91.2% of NCC-positive cells and only in NCC-positive cells, revealing high recombination efficiency and DCT specificity. Crossing to R26R-TdTomato mice revealed higher leakiness (64.5%), suggesting differential sensitivity of the floxed site. Western blot analysis revealed no differences in abundances of total NCC or the active phosphorylated form of NCC in Slc12a3-Cre-ERT2 mice of either sex compared with controls. Plasma K+ and Mg2+ concentrations and thiazide-sensitive Na+ and K+ excretion did not differ in Slc12a3-Cre-ERT2 mice compared with controls when sex matched. These data suggest genetic modification had no obvious effect on NCC function. Slc12a3-Cre-ERT2 mice are the first line generated demonstrating inducible Cre recombinase activity along the entire DCT and will be a useful tool to study DCT function.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Xiao-Tong Su
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jin-Jin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad California Institute of Regenerative Medicine Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad California Institute of Regenerative Medicine Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon.,Veterans Affairs Portland Health Care System, Portland, Oregon
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad California Institute of Regenerative Medicine Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
29
|
Bi Y, Li C, Zhang Y, Wang Y, Chen S, Yue Q, Hoover RS, Wang XH, Delpire E, Eaton DC, Zhuang J, Cai H. Stimulatory Role of SPAK Signaling in the Regulation of Large Conductance Ca 2+-Activated Potassium (BK) Channel Protein Expression in Kidney. Front Physiol 2020; 11:638. [PMID: 32714200 PMCID: PMC7343913 DOI: 10.3389/fphys.2020.00638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/20/2020] [Indexed: 12/30/2022] Open
Abstract
SPS1-related proline/alanine-rich kinase (SPAK) plays important roles in regulating the function of numerous ion channels and transporters. With-no-lysine (WNK) kinase phosphorylates SPAK kinase to active the SPAK signaling pathway. Our previous studies indicated that WNK kinases regulate the activity of the large-conductance Ca2+-activated K+ (BK) channel and its protein expression via the ERK1/2 signaling pathway. It remains largely unknown whether SPAK kinase directly modulates the BK protein expression in kidney. In this study, we investigated the effect of SPAK on renal BK protein expression in both HEK293 cells and mouse kidney. In HEK293 cells, siRNA-mediated knockdown of SPAK expression significantly reduced BK protein expression and increased ERK1/2 phosphorylation, whereas overexpression of SPAK significantly enhanced BK expression and decreased ERK1/2 phosphorylation in a dose-dependent manner. Knockdown of ERK1/2 prevented SPAK siRNA-mediated inhibition of BK expression. Similarly, pretreatment of HEK293 cells with either the lysosomal inhibitor bafilomycin A1 or the proteasomal inhibitor MG132 reversed the inhibitory effects of SPAK knockdown on BK expression. We also found that there is no BK channel activity in PCs of CCD in SPAK KO mice using the isolated split-open tubule single-cell patching. In addition, we found that BK protein abundance in the kidney of SPAK knockout mice was significantly decreased and ERK1/2 phosphorylation was significantly enhanced. A high-potassium diet significantly increased BK protein abundance and SPAK phosphorylation levels, while reducing ERK1/2 phosphorylation levels. These findings suggest that SPAK enhances BK protein expression by reducing ERK1/2 signaling-mediated lysosomal and proteasomal degradations of the BK channel.
Collapse
Affiliation(s)
- Ye Bi
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China.,Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Chunmei Li
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Yiqian Zhang
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunman Wang
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Shan Chen
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Qiang Yue
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Robert S Hoover
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
| | - Xiaonan H Wang
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, TN, United States
| | - Douglas C Eaton
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | - Jieqiu Zhuang
- Department of Pediatric Nephrology, The Second Affiliated Hospital/Yuying Children Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui Cai
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States.,Section of Nephrology, Atlanta Veterans Administration Medical Center, Decatur, GA, United States
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The apical Na/K/2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb, contributing to maintenance of blood pressure (BP). Despite effective NKCC2 inhibition by loop diuretics, these agents are not viable for long-term management of BP due to side effects. Novel molecular mechanisms that control NKCC2 activity reveal an increasingly complex picture with interacting layers of NKCC2 regulation. Here, we review the latest developments that shine new light on NKCC2-mediated control of BP and potential new long-term therapies to treat hypertension. RECENT FINDINGS Emerging molecular NKCC2 regulators, often binding partners, reveal a complex overlay of interacting mechanisms aimed at fine tuning NKCC2 activity. Different factors achieve this by shifting the balance between trafficking steps like exocytosis, endocytosis, recycling and protein turnover, or by balancing phosphorylation vs. dephosphorylation. Further molecular details are also emerging on previously known pathways of NKCC2 regulation, and recent in-vivo data continues to place NKCC2 regulation at the center of BP control. SUMMARY Several layers of emerging molecular mechanisms that control NKCC2 activity may operate simultaneously, but they can also be controlled independently. This provides an opportunity to identify new pharmacological targets to fine-tune NKCC2 activity for BP management.
Collapse
|
31
|
De novo variants in CUL3 are associated with global developmental delays with or without infantile spasms. J Hum Genet 2020; 65:727-734. [PMID: 32341456 DOI: 10.1038/s10038-020-0758-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/16/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022]
Abstract
The ubiquitin-proteasome system is the principal system for protein degradation mediated by ubiquitination and is involved in various cellular processes. Cullin-RING ligases (CRL) are one class of E3 ubiquitin ligases that mediate polyubiquitination of specific target proteins, leading to decomposition of the substrate. Cullin 3 (CUL3) is a member of the Cullin family proteins, which act as scaffolds of CRL. Here we describe three cases of global developmental delays, with or without epilepsy, who had de novo CUL3 variants. One missense variant c.854T>C, p.(Val285Ala) and two frameshift variants c.137delG, p.(Arg46Leufs*32) and c.1239del, p.(Asp413Glufs*42) were identified by whole-exome sequencing. The Val285 residue located in the Cullin N-terminal domain and p.Val285Ala CUL3 mutant showed significantly weaker interactions to the BTB domain proteins than wild-type CUL3. Our findings suggest that de novo CUL3 variants may cause structural instability of the CRL complex and impairment of the ubiquitin-proteasome system, leading to diverse neuropsychiatric disorders.
Collapse
|
32
|
Furusho T, Uchida S, Sohara E. The WNK signaling pathway and salt-sensitive hypertension. Hypertens Res 2020; 43:733-743. [PMID: 32286498 DOI: 10.1038/s41440-020-0437-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022]
Abstract
The distal nephron of the kidney has a central role in sodium and fluid homeostasis, and disruption of this homeostasis due to mutations of with-no-lysine kinase 1 (WNK1), WNK4, Kelch-like 3 (KLHL3), or Cullin 3 (CUL3) causes pseudohypoaldosteronism type II (PHAII), an inherited hypertensive disease. WNK1 and WNK4 activate the NaCl cotransporter (NCC) at the distal convoluted tubule through oxidative stress-responsive gene 1 (OSR1)/Ste20-related proline-alanine-rich kinase (SPAK), constituting the WNK-OSR1/SPAK-NCC phosphorylation cascade. The level of WNK protein is regulated through degradation by the CUL3-KLHL3 E3 ligase complex. In the normal state, the activity of WNK signaling in the kidney is physiologically regulated by sodium intake to maintain sodium homeostasis in the body. In patients with PHAII, however, because of the defective degradation of WNK kinases, NCC is constitutively active and not properly suppressed by a high salt diet, leading to abnormally increased salt reabsorption and salt-sensitive hypertension. Importantly, recent studies have demonstrated that potassium intake, insulin, and TNFα are also physiological regulators of WNK signaling, suggesting that they contribute to the salt-sensitive hypertension associated with a low potassium diet, metabolic syndrome, and chronic kidney disease, respectively. Moreover, emerging evidence suggests that WNK signaling also has some unique roles in metabolic, cardiovascular, and immunological organs. Here, we review the recent literature and discuss the molecular mechanisms of the WNK signaling pathway and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Taisuke Furusho
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
33
|
Ostrosky-Frid M, Chávez-Canales M, Romo M, Grunfeld B, Simsolo R, Segura-Kato Y, Tusié-Luna T, Gamba G. Familial Hyperkalemic Hypertension Genotype With a Negative Phenotype: A CUL3 Mosaicism. Am J Hypertens 2020; 33:278-281. [PMID: 31802109 DOI: 10.1093/ajh/hpz185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/21/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mauricio Ostrosky-Frid
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
- PECEM (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
| | - María Chávez-Canales
- Unidad de Medicina Traslacional, Instituto Nacional de Cardiología Ignacio Chávez e Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Miriam Romo
- Consultorio de Hipertensión Arterial del Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Beatriz Grunfeld
- Consultorio de Hipertensión Arterial del Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Rosa Simsolo
- Consultorio de Hipertensión Arterial del Hospital de Niños “Ricardo Gutiérrez”, Buenos Aires, Argentina
| | - Yayoi Segura-Kato
- Molecular Biology and Genomic Medicine Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
- Molecular Physiology Unit and Department of Genomic Medicine and Enviromental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - Teresa Tusié-Luna
- Molecular Biology and Genomic Medicine Unit, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
- Molecular Physiology Unit and Department of Genomic Medicine and Enviromental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
| | - Gerardo Gamba
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Mexico City, Mexico
- PECEM (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacan, Mexico City, Mexico
- Molecular Physiology Unit and Department of Genomic Medicine and Enviromental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Tlalpan, Mexico City, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, NL, Mexico
| |
Collapse
|
34
|
Hoorn EJ, Gritter M, Cuevas CA, Fenton RA. Regulation of the Renal NaCl Cotransporter and Its Role in Potassium Homeostasis. Physiol Rev 2020; 100:321-356. [PMID: 31793845 DOI: 10.1152/physrev.00044.2018] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Daily dietary potassium (K+) intake may be as large as the extracellular K+ pool. To avoid acute hyperkalemia, rapid removal of K+ from the extracellular space is essential. This is achieved by translocating K+ into cells and increasing urinary K+ excretion. Emerging data now indicate that the renal thiazide-sensitive NaCl cotransporter (NCC) is critically involved in this homeostatic kaliuretic response. This suggests that the early distal convoluted tubule (DCT) is a K+ sensor that can modify sodium (Na+) delivery to downstream segments to promote or limit K+ secretion. K+ sensing is mediated by the basolateral K+ channels Kir4.1/5.1, a capacity that the DCT likely shares with other nephron segments. Thus, next to K+-induced aldosterone secretion, K+ sensing by renal epithelial cells represents a second feedback mechanism to control K+ balance. NCC’s role in K+ homeostasis has both physiological and pathophysiological implications. During hypovolemia, NCC activation by the renin-angiotensin system stimulates Na+ reabsorption while preventing K+ secretion. Conversely, NCC inactivation by high dietary K+ intake maximizes kaliuresis and limits Na+ retention, despite high aldosterone levels. NCC activation by a low-K+ diet contributes to salt-sensitive hypertension. K+-induced natriuresis through NCC offers a novel explanation for the antihypertensive effects of a high-K+ diet. A possible role for K+ in chronic kidney disease is also emerging, as epidemiological data reveal associations between higher urinary K+ excretion and improved renal outcomes. This comprehensive review will embed these novel insights on NCC regulation into existing concepts of K+ homeostasis in health and disease.
Collapse
Affiliation(s)
- Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Martin Gritter
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Catherina A. Cuevas
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Robert A. Fenton
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands; and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
35
|
Cornelius RJ, Yang CL, Ellison DH. Hypertension-causing cullin 3 mutations disrupt COP9 signalosome binding. Am J Physiol Renal Physiol 2019; 318:F204-F208. [PMID: 31813255 DOI: 10.1152/ajprenal.00497.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The discovery of new genetic mutations that cause hypertension has illuminated previously unrecognized physiological pathways. One such regulatory pathway was identified when mutations in with no lysine kinase (WNK)4, Kelch-like 3 (KLHL3), and cullin 3 (CUL3) were shown to cause the disease familial hyperkalemic hypertension (FHHt). Mutations in all three genes upregulate the NaCl cotransporter (NCC) due to an impaired ability to degrade WNK protein through the cullin-RING-ligase (CRL) ubiquitin-proteasome system. The CUL3 FHHt mutations cause the most severe phenotype, yet the precise mechanism by which these mutations cause the disease has not been established and current proposed models are controversial. New data have identified a possible novel mechanism involving dysregulation of CUL3 activity by the COP9 signalosome (CSN). The CSN interaction with mutant CUL3 is diminished, causing hyperneddylation of the CRL. Recent work has shown that direct renal CSN impairment mimics some aspects of the CUL3 mutation, including lower KLHL3 abundance and activation of the WNK-NCC pathway. Furthermore, in vitro and in vivo studies of CSN inhibition have shown selective degradation of CRL substrate adaptors via auto-ubiquitination, allowing substrate accumulation. In this review, we will focus on recent research that highlights the role of the CSN role in CUL3 mutations that cause FHHt. We will also highlight how these results inform other recent studies of CSN dysfunction.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon.,Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
36
|
Ferdaus MZ, Mukherjee A, Nelson JW, Blatt PJ, Miller LN, Terker AS, Staub O, Lin DH, McCormick JA. Mg 2+ restriction downregulates NCC through NEDD4-2 and prevents its activation by hypokalemia. Am J Physiol Renal Physiol 2019; 317:F825-F838. [PMID: 31364380 PMCID: PMC6843039 DOI: 10.1152/ajprenal.00216.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Hypomagnesemia is associated with reduced kidney function and life-threatening complications and sustains hypokalemia. The distal convoluted tubule (DCT) determines final urinary Mg2+ excretion and, via activity of the Na+-Cl- cotransporter (NCC), also plays a key role in K+ homeostasis by metering Na+ delivery to distal segments. Little is known about the mechanisms by which plasma Mg2+ concentration regulates NCC activity and how low-plasma Mg2+ concentration and K+ concentration interact to modulate NCC activity. To address this, we performed dietary manipulation studies in mice. Compared with normal diet, abundances of total NCC and phosphorylated NCC (pNCC) were lower after short-term (3 days) or long-term (14 days) dietary Mg2+ restriction. Altered NCC activation is unlikely to play a role, since we also observed lower total NCC abundance in mice lacking the two NCC-activating kinases, STE20/SPS-1-related proline/alanine-rich kinase and oxidative stress response kinase-1, after Mg2+ restriction. The E3 ubiquitin-protein ligase NEDD4-2 regulates NCC abundance during dietary NaCl loading or K+ restriction. Mg2+ restriction did not lower total NCC abundance in inducible nephron-specific neuronal precursor cell developmentally downregulated 4-2 (NEDD4-2) knockout mice. Total NCC and pNCC abundances were similar after short-term Mg2+ or combined Mg2+-K+ restriction but were dramatically lower compared with a low-K+ diet. Therefore, sustained NCC downregulation may serve a mechanism that enhances distal Na+ delivery during states of hypomagnesemia, maintaining hypokalemia. Similar results were obtained with long-term Mg2+-K+ restriction, but, surprisingly, NCC was not activated after long-term K+ restriction despite lower plasma K+ concentration, suggesting significant differences in distal tubule adaptation to acute or chronic K+ restriction.
Collapse
Affiliation(s)
- Mohammed Z. Ferdaus
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Anindit Mukherjee
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W. Nelson
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Philip J. Blatt
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Lauren N. Miller
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Andrew S. Terker
- 2Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Olivier Staub
- 3Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Dao-Hong Lin
- 4Department of Pharmacology, New York Medical College, Valhalla, New York
| | - James A. McCormick
- 1Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
37
|
Cornelius RJ, Ferdaus MZ, Nelson JW, McCormick JA. Cullin-Ring ubiquitin ligases in kidney health and disease. Curr Opin Nephrol Hypertens 2019; 28:490-497. [PMID: 31313673 PMCID: PMC6686897 DOI: 10.1097/mnh.0000000000000527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Members of the Cullin family act as scaffolds in E3 ubiquitin ligases and play a central role in mediating protein degradation. Interactions with many different substrate-binding adaptors permit Cullin-containing E3 ligases to participate in diverse cellular functions. In the kidney, one well established target of Cullin-mediated degradation is the transcription factor Nrf2, a key player in responses to oxidative stress. The goal of this review is to discuss more recent findings revealing broader roles for Cullins in the kidney. RECENT FINDINGS Cullin 3 acts as the scaffold in the E3 ligase regulating Nrf2 abundance, but was more recently shown to be mutated in the disease familial hyperkalemic hypertension. Studies seeking to elucidate the molecular mechanisms by which Cullin 3 mutations lead to dysregulation of renal sodium transport will be discussed. Disruption of Cullin 3 in mice unexpectedly causes polyuria and fibrotic injury suggesting it has additional roles in the kidney. We will also review recent transcriptomic data suggesting that other Cullins are also likely to play important roles in renal function. SUMMARY Cullins form a large and diverse family of E3 ubiquitin ligases that are likely to have many important functions in the kidney.
Collapse
Affiliation(s)
- Ryan J. Cornelius
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR
| | - Mohammed Z. Ferdaus
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR
| | - Jonathan W. Nelson
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR
| | - James A. McCormick
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, OR
| |
Collapse
|
38
|
Guo DF, Rahmouni K. The Bardet-Biedl syndrome protein complex regulates cell migration and tissue repair through a Cullin-3/RhoA pathway. Am J Physiol Cell Physiol 2019; 317:C457-C465. [PMID: 31216194 DOI: 10.1152/ajpcell.00498.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cell motility and migration play critical roles in various physiological processes and disease states. Here, we show that the BBBsome, a macromolecule composed of eight Bardet-Biedl syndrome (BBS) proteins including BBS1, is a critical determinant of cell migration and wound healing. Fibroblast cells derived from mice or humans harboring a homozygous missense mutation (BBS1M390R/M390R) that disrupt the BBSome exhibit defects in migration and wound healing. Furthermore, we demonstrate that BBS1M390R/M390R mice have significantly delayed wound closure. In line with this, we provide data suggesting that BBS1M390R/M390R fibroblasts have impaired platelet-derived growth factor-AA (PDGF) receptor-α signaling, a key regulator of directional cell migration acting as a chemoattractant during postnatal migration responses such as wound healing. In addition, we show that BBS1M390R/M390R fibroblasts have upregulated RhoA expression and activity. The relevance of RhoA upregulation is demonstrated by the ability of RhoA-kinase inhibitor Y27632 to partially rescue the migration defect of BBS1M390R/M390R fibroblasts cells. We also show that accumulation of RhoA protein in BBS1M390R/M390R fibroblasts cells is associated with reduction and inactivation of the ubiquitin ligase Cullin-3. Consistent with this, Cullin-3 inhibition with MLN4924 is sufficient to reduce migration of normal fibroblasts. These data implicate the BBSome in cell motility and tissue repair through a mechanism that involves PDGF receptor signaling and Cullin-3-mediated control of RhoA.
Collapse
Affiliation(s)
- Deng-Fu Guo
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Obesity Education and Research Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Fraternal Order of Eagles Diabetes Research, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Veterans Affairs Health Care System, Iowa City, Iowa
| |
Collapse
|
39
|
Agbor LN, Nair AR, Wu J, Lu KT, Davis DR, Keen HL, Quelle FW, McCormick JA, Singer JD, Sigmund CD. Conditional deletion of smooth muscle Cullin-3 causes severe progressive hypertension. JCI Insight 2019; 5:129793. [PMID: 31184598 DOI: 10.1172/jci.insight.129793] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.
Collapse
Affiliation(s)
- Larry N Agbor
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Anand R Nair
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jing Wu
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ko-Ting Lu
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Deborah R Davis
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Henry L Keen
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Frederick W Quelle
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - James A McCormick
- Division of Nephrology & Hypertension, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Jeffrey D Singer
- Department of Biology, Portland State University, Portland, Oregon, USA
| | - Curt D Sigmund
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
40
|
Abdel Khalek W, Rafael C, Loisel-Ferreira I, Kouranti I, Clauser E, Hadchouel J, Jeunemaitre X. Severe Arterial Hypertension from Cullin 3 Mutations Is Caused by Both Renal and Vascular Effects. J Am Soc Nephrol 2019; 30:811-823. [PMID: 30967423 DOI: 10.1681/asn.2017121307] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/27/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Mutations in four genes, WNK lysine deficient protein kinase 1 and 4 (WNK1 and WNK4), kelch like family member 3 (KLHL3), or Cullin 3 (CUL3), can result in familial hyperkalemic hypertension (FHHt), a rare Mendelian form of human arterial hypertension. Although all mutations result in an increased abundance of WNK1 or WNK4, all FHHt-causing CUL3 mutations, resulting in the skipping of exon 9, lead to a more severe phenotype. METHODS We created and compared two mouse models, one expressing the mutant Cul3 protein ubiquitously (pgk-Cul3∆9) and the other specifically in vascular smooth muscle cells (SM22-Cul3∆9). We conducted pharmacologic investigations on isolated aortas and generated stable and inducible HEK293 cell lines that overexpress the wild-type Cul3 or mutant Cul3 (Cul3∆9) protein. RESULTS As expected, pgk-Cul3∆9 mice showed marked hypertension with significant hyperkalemia, hyperchloremia and low renin. BP increased significantly in SM22-Cul3∆9 mice, independent of any measurable effect on renal transport. Only pgk-Cul3∆9 mice displayed increased expression of the sodium chloride cotransporter and phosphorylation by the WNK-SPAK kinases. Both models showed altered reactivity of isolated aortas to phenylephrine and acetylcholine, as well as marked acute BP sensitivity to the calcium channel blocker amlodipine. Aortas from SM22-Cul3∆9 mice showed increased expression of RhoA, a key molecule involved in regulation of vascular tone, compared with aortas from control mice. We also observed increased RhoA abundance and t 1/2 in Cul3∆9-expressing cells, caused by decreased ubiquitination. CONCLUSIONS Mutations in Cul3 cause severe hypertension by affecting both renal and vascular function, the latter being associated with activation of RhoA.
Collapse
Affiliation(s)
- Waed Abdel Khalek
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France.,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Chloé Rafael
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France.,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale UMR_S1155, Tenon Hospital, Paris, France.,Faculty of Medicine, University Pierre and Marie Curie, Paris, France.,Faculty of Sciences, University Paris-Diderot, Sorbonne Paris Cité, Paris, France; and
| | - Irmine Loisel-Ferreira
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France.,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Ilektra Kouranti
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France.,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Eric Clauser
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France.,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France
| | - Juliette Hadchouel
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France; .,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale UMR_S1155, Tenon Hospital, Paris, France.,Faculty of Medicine, University Pierre and Marie Curie, Paris, France
| | - Xavier Jeunemaitre
- Institut National de la Santé et de la Recherche Médicale U970, Paris Cardiovascular Research Center, Paris, France; .,Faculty of Medicine, University Paris-Descartes, Sorbonne Paris Cité, Paris, France.,Department of Genetics, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
41
|
Disruption of CUL3-mediated ubiquitination causes proximal tubule injury and kidney fibrosis. Sci Rep 2019; 9:4596. [PMID: 30872636 PMCID: PMC6418206 DOI: 10.1038/s41598-019-40795-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cullin 3 (CUL3) is part of the ubiquitin proteasomal system and controls several cellular processes critical for normal organ function including the cell cycle, and Keap1/Nrf2 signaling. Kidney tubule-specific Cul3 disruption causes tubulointerstitial fibrosis, but little is known about the mechanisms. Therefore, we tested the hypothesis that dysregulation of the cell cycle and Keap1/Nrf2 pathway play a role in initiating the kidney injury upon Cul3 disruption. Cul3 deletion increased expression of cyclin E and p21, associated with uncontrolled proliferation, DNA damage, and apoptosis, all of which preceded proximal tubule injury. The cdk2-cyclin E inhibitor roscovitine did not prevent the effects of Cul3 deletion, but instead exacerbated the kidney injury. Injury occurred despite accumulation and activation of CUL3 substrate Keap1/Nrf2, proposed to be protective in kidney injury. Cul3 disruption led to progressive interstitial inflammation, functionally relevant renal fibrosis and death. Finally, we observed reduced CUL3 expression in several AKI and CKD mouse models and in fibrotic human kidney tissue. These data establish CUL3 knockout mice as a novel genetic CKD model in which dysregulation of the cell cycle may play a primary role in initiating tubule injury, and that CUL3 dysregulation could contribute to acute and fibrotic kidney disease.
Collapse
|
42
|
Cornelius RJ, Si J, Cuevas CA, Nelson JW, Gratreak BDK, Pardi R, Yang CL, Ellison DH. Renal COP9 Signalosome Deficiency Alters CUL3-KLHL3-WNK Signaling Pathway. J Am Soc Nephrol 2018; 29:2627-2640. [PMID: 30301860 PMCID: PMC6218864 DOI: 10.1681/asn.2018030333] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 09/07/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The familial hyperkalemic hypertension (FHHt) cullin 3 (CUL3) mutant does not degrade WNK kinases normally, thereby leading to thiazide-sensitive Na-Cl cotransporter (NCC) activation. CUL3 mutant (CUL3Δ9) does not bind normally to the COP9 signalosome (CSN), a deneddylase involved in regulating cullin-RING ligases. CUL3Δ9 also caused increased degradation of the CUL3-WNK substrate adaptor kelch-like 3 (KLHL3). Here, we sought to determine how defective CSN action contributes to the CUL3Δ9 phenotype. METHODS The Pax8/LC1 mouse system was used to generate mice in which the catalytically active CSN subunit, Jab1, was deleted only along the nephron, after full development (KS-Jab1-/-). RESULTS Western blot analysis demonstrated that Jab1 deletion increased the abundance of neddylated CUL3. Moreover, total CUL3 expression was reduced, suggesting decreased CUL3 stability. KLHL3 was almost completely absent in KS-Jab1-/- mice. Conversely, the protein abundances of WNK1, WNK4, and SPAK kinases were substantially higher. Activation of WNK4, SPAK, and OSR1 was indicated by higher phosphorylated protein levels and translocation of the proteins into puncta, as observed by immunofluorescence. The ratio of phosphorylated NCC to total NCC was also higher. Surprisingly, NCC protein abundance was low, likely contributing to hypokalemia and Na+ and K+ wasting. Additionally, long-term Jab1 deletion resulted in kidney damage. CONCLUSIONS Together, the results indicate that deficient CSN binding contributes importantly to the FHHt phenotype. Although defective CUL3Δ9-faciliated WNK4 degradation likely contributes, dominant effects on KLHL3 may be a second factor that is necessary for the phenotype.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jinge Si
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Catherina A Cuevas
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Brittany D K Gratreak
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - Ruggero Pardi
- School of Medicine and Scientific Institute, San Raffaele University, Milan, Italy; and
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon;
- Renal Section, Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
43
|
Cornelius RJ, Zhang C, Erspamer KJ, Agbor LN, Sigmund CD, Singer JD, Yang CL, Ellison DH. Dual gain and loss of cullin 3 function mediates familial hyperkalemic hypertension. Am J Physiol Renal Physiol 2018; 315:F1006-F1018. [PMID: 29897280 PMCID: PMC6230741 DOI: 10.1152/ajprenal.00602.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/11/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022] Open
Abstract
Familial hyperkalemic hypertension is caused by mutations in with-no-lysine kinases (WNKs) or in proteins that mediate their degradation, kelch-like 3 (KLHL3) and cullin 3 (CUL3). Although the mechanisms by which WNK and KLHL3 mutations cause the disease are now clear, the effects of the disease-causing CUL3Δ403-459 mutation remain controversial. Possible mechanisms, including hyperneddylation, altered ubiquitin ligase activity, decreased association with the COP9 signalosome (CSN), and increased association with and degradation of KLHL3 have all been postulated. Here, we systematically evaluated the effects of Cul3Δ403-459 using cultured kidney cells. We first identified that the catalytically active CSN subunit jun activation domain-binding protein-1 (JAB1) does not associate with the deleted Cul3 4-helix bundle domain but instead with the adjacent α/β1 domain, suggesting that altered protein folding underlies the impaired binding. Inhibition of deneddylation with JAB1 siRNA increased Cul3 neddylation and decreased KLHL3 abundance, similar to the Cul3 mutant. We next determined that KLHL3 degradation has both ubiquitin ligase-dependent and -independent components. Proteasomal KLHL3 degradation was enhanced by Cul3Δ403-459; however, autophagic degradation was also upregulated by this Cul3 mutant. Finally, to evaluate whether deficient substrate adaptor was responsible for the disease, we restored KLHL3 to wild-type (WT) Cul3 levels. In the absence of WT Cul3, WNK4 was not degraded, demonstrating that Cul3Δ403-459 itself cannot degrade WNK4; conversely, when WT Cul3 was present, as in diseased humans, WNK4 degradation was restored. In conclusion, deletion of exon 9 from Cul3 generates a protein that is itself ubiquitin-ligase defective but also capable of enhanced autophagocytic KLHL3 degradation, thereby exerting dominant-negative effects on the WT allele.
Collapse
Affiliation(s)
- Ryan J Cornelius
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
| | - Chong Zhang
- Department of Nephrology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Kayla J Erspamer
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
| | - Larry N Agbor
- Department of Pharmacology, UIHC Center for Hypertension Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Curt D Sigmund
- Department of Pharmacology, UIHC Center for Hypertension Research, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Jeffrey D Singer
- Department of Biology, Portland State University , Portland, Oregon
| | - Chao-Ling Yang
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
| | - David H Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
- Veterans Affairs Portland Health Care System, Portland, Oregon
| |
Collapse
|
44
|
Ostrosky-Frid M, Castañeda-Bueno M, Gamba G. Regulation of the renal NaCl cotransporter by the WNK/SPAK pathway: lessons learned from genetically altered animals. Am J Physiol Renal Physiol 2018; 316:F146-F158. [PMID: 30089030 DOI: 10.1152/ajprenal.00288.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The renal thiazide-sensitive NaCl cotransporter (NCC) is the major salt transport pathway in the distal convoluted tubule of the mammalian nephron. NCC activity is critical for modulation of arterial blood pressure and serum potassium levels. Reduced activity of NCC in genetic diseases results in arterial hypotension and hypokalemia, while increased activity results in genetic diseases featuring hypertension and hyperkalemia. Several hormones and physiological conditions modulate NCC activity through a final intracellular complex pathway involving kinases and ubiquitin ligases. A substantial amount of work has been conducted to understand this pathway in the last 15 yr, but advances over the last 3 yr have helped to begin to understand how these regulatory proteins interact with each other and modulate the activity of this important cotransporter. In this review, we present the current model of NCC regulation by the Cullin 3 protein/Kelch-like 3 protein/with no lysine kinase/STE20-serine-proline alanine-rich kinase (CUL3/KELCH3-WNK-SPAK) pathway. We present a review of all genetically altered mice that have been used to translate most of the proposals made from in vitro experiments into in vivo observations that have helped to elucidate the model at the physiological level. Many questions have been resolved, but some others will require further models to be constructed. In addition, unexpected observations in mice have raised new questions and identified regulatory pathways that were previously unknown.
Collapse
Affiliation(s)
- Mauricio Ostrosky-Frid
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Mexico City, Mexico.,PECEM, Facultad de Medicina, Universidad Nacional Autónoma de México , Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México , Mexico City, Mexico.,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , Mexico City, Mexico.,Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| |
Collapse
|
45
|
Yoshida S, Araki Y, Mori T, Sasaki E, Kasagi Y, Isobe K, Susa K, Inoue Y, Bomont P, Okado T, Rai T, Uchida S, Sohara E. Decreased KLHL3 expression is involved in the pathogenesis of pseudohypoaldosteronism type II caused by cullin 3 mutation in vivo. Clin Exp Nephrol 2018; 22:1251-1257. [PMID: 29869755 DOI: 10.1007/s10157-018-1593-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/25/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pseudohypoaldosteronism type II (PHAII) is a hereditary hypertensive disease caused by mutations in four genes: WNK1, WNK4, Kelch-like3 (KLHL3), and cullin3 (CUL3). Recently, it was revealed that CUL3-KLHL3 E3 ligase complex ubiquitinates WNK1 and WNK4, leading to their degradation, and that a common pathogenesis of PHAII is defective WNK degradation due to CUL3-KLHL3 E3 ligase complex impairment. PHAII-causing CUL3 mutations mediate exon9 skipping, producing a CUL3 protein with a 57-amino acid deletion (Δ403-459). However, the pathogenic effects of KLHL3, an adaptor protein that links WNKs with CUL3, in PHAII caused by CUL3 mutation remain unclear. METHODS To clarify detailed pathophysiological mechanisms underlying PHAII caused by CUL3 mutation in vivo, we generated and analyzed knock-in mice carrying the same CUL3 exon9 deletion (CUL3WT/Δex9) as that reported in PHAII patients. RESULTS CUL3WT/Δex9 mice exhibited a PHAII-like phenotype. Interestingly, we confirmed markedly decreased KLHL3 expression in CUL3WT/Δex9 mice by confirming the true KLHL3 band in vivo. However, the expression of other KLHL family proteins, such as KLHL2, was comparable between WT and mutant mice. CONCLUSION KLHL3 expression was decreased in CUL3WT/Δex9 mice. However, expression levels of other KLHL family proteins were comparable between the wild-type and mutant mice. These findings indicate that the decreased abundance of KLHL3 is a specific phenomenon caused by mutant CUL3 (Δexon9). Our findings would improve our understanding of the pathogenesis of PHAII caused by CUL3 mutation in vivo.
Collapse
Affiliation(s)
- Sayaka Yoshida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Yuya Araki
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Takayasu Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Emi Sasaki
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Yuri Kasagi
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Kiyoshi Isobe
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Koichiro Susa
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Yuichi Inoue
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Pascale Bomont
- Avenir-Atip Team, INM, INSERM, University of Montpellier, 34091, Montpellier Cedex 5, France
| | - Tomokazu Okado
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Tatemitsu Rai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo, 113-8519, Japan.
| |
Collapse
|
46
|
Ferdaus MZ, McCormick JA. Mechanisms and controversies in mutant Cul3-mediated familial hyperkalemic hypertension. Am J Physiol Renal Physiol 2018; 314:F915-F920. [PMID: 29361671 DOI: 10.1152/ajprenal.00593.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Autosomal dominant mutations in cullin-3 ( Cul3) cause the most severe form of familial hyperkalemic hypertension (FHHt). Cul3 mutations cause skipping of exon 9, which results in an internal deletion of 57 amino acids from the CUL3 protein (CUL3-∆9). The precise mechanism by which this altered form of CUL3 causes FHHt is controversial. CUL3 is a member of the cullin-RING ubiquitin ligase family that mediates ubiquitination and thus degradation of cellular proteins, including with-no-lysine [K] kinases (WNKs). In CUL3-∆9-mediated FHHt, proteasomal degradation of WNKs is abrogated, leading to overactivation of the WNK targets sterile 20/SPS-1 related proline/alanine-rich kinase and oxidative stress-response kinase-1, which directly phosphorylate and activate the thiazide-sensitive Na+-Cl- cotransporter. Several groups have suggested different mechanisms by which CUL3-∆9 causes FHHt. The majority of these are derived from in vitro data, but recently the Kurz group (Schumacher FR, Siew K, Zhang J, Johnson C, Wood N, Cleary SE, Al Maskari RS, Ferryman JT, Hardege I, Figg NL, Enchev R, Knebel A, O'Shaughnessy KM, Kurz T. EMBO Mol Med 7: 1285-1306, 2015) described the first mouse model of CUL3-∆9-mediated FHHt. Analysis of this model suggested that CUL3-∆9 is degraded in vivo, and thus Cul3 mutations cause FHHt by inducing haploinsufficiency. We recently directly tested this model but found that other dominant effects of CUL3-∆9 must contribute to the development of FHHt. In this review, we focus on our current knowledge of CUL3-∆9 action gained from in vitro and in vivo models that may help unravel this complex problem.
Collapse
Affiliation(s)
- Mohammed Z Ferdaus
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University , Portland, Oregon
| |
Collapse
|