1
|
Sun X, Yegambaram M, Lu Q, Garcia Flores AE, Pokharel MD, Soto J, Aggarwal S, Wang T, Fineman JR, Black SM. Mitochondrial fission produces a Warburg effect via the oxidative inhibition of prolyl hydroxylase domain-2. Redox Biol 2025; 81:103529. [PMID: 39978304 PMCID: PMC11889635 DOI: 10.1016/j.redox.2025.103529] [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: 12/19/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Excessive mitochondrial fission and a shift to a Warburg phenotype are hallmarks of pulmonary hypertension (PH), although the mechanistic link between these processes remains unclear. We show that in pulmonary arterial endothelial cells (PAEC), Drp1 overexpression induces mitochondrial fission and increases glycolytic ATP production and glycolysis. This is due to mitochondrial reactive oxygen species (mito-ROS)-mediated activation of hypoxia-inducible factor-1α (HIF-1α) signaling, and this is linked to hydrogen peroxide (H2O2)-mediated inhibition of prolyl hydroxylase domain-2 (PHD2) due to its cysteine 326 oxidation and dimerization. Furthermore, these findings are validated in PAEC isolated from a lamb model of PH, which are glycolytic (Shunt PAEC), exhibit increases in both H2O2 and PHD2 dimer levels. The overexpression of catalase reversed the PHD2 dimerization, decreased HIF-1α levels, and attenuated glycolysis in Shunt PAEC. Our data suggest that reducing PHD2 dimerization could be a potential therapeutic target for PH.
Collapse
Affiliation(s)
- Xutong Sun
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Manivannan Yegambaram
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Qing Lu
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Alejandro E Garcia Flores
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Marissa D Pokharel
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jamie Soto
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA
| | - Saurabh Aggarwal
- The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jeffrey R Fineman
- The Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; The Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Florida International University, Center for Translational Science, Port Saint Lucie, FL, 34987, USA; The Departments of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; The Departments of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
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
|
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
|
4
|
Staels B, Butruille L, Francque S. Treating NASH by targeting peroxisome proliferator-activated receptors. J Hepatol 2023; 79:1302-1316. [PMID: 37459921 DOI: 10.1016/j.jhep.2023.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/18/2023] [Accepted: 07/02/2023] [Indexed: 09/15/2023]
Abstract
The pathophysiology of non-alcoholic steatohepatitis (NASH) encompasses a complex set of intra- and extrahepatic driving mechanisms, involving numerous metabolic, inflammatory, vascular and fibrogenic pathways. The peroxisome proliferator-activated receptors (PPARs) α, β/δ and γ belong to the nuclear receptor family of ligand-activated transcription factors. Activated PPARs modulate target tissue transcriptomic profiles, enabling the body's adaptation to changing nutritional, metabolic and inflammatory environments. PPARs hence regulate several pathways involved in NASH pathogenesis. Whereas single PPAR agonists exert robust anti-NASH activity in several preclinical models, their clinical effects on histological endpoints of NASH resolution and fibrosis regression appear more modest. Simultaneous activation of several PPAR isotypes across different organs and within-organ cell types, resulting in pleiotropic actions, enhances the therapeutic potential of PPAR agonists as pharmacological agents for NASH and NASH-related hepatic and extrahepatic morbidity, with some compounds having already shown clinical efficacy on histological endpoints.
Collapse
Affiliation(s)
- Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Laura Butruille
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, B-2650, Edegem, Belgium; InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Belgium.
| |
Collapse
|
5
|
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: 2] [Impact Index Per Article: 1.0] [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
|
6
|
Kumar G, Fang S, Golosova D, Lu KT, Brozoski DT, Vazirabad I, Sigmund CD. Structure and Function of RhoBTB1 Required for Substrate Specificity and Cullin-3 Ubiquitination. FUNCTION 2023; 4:zqad034. [PMID: 37575477 PMCID: PMC10413933 DOI: 10.1093/function/zqad034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 08/15/2023] Open
Abstract
We identified Rho-related BTB domain containing 1 (RhoBTB1) as a key regulator of phosphodiesterase 5 (PDE5) activity, and through PDE5, a regulator of vascular tone. We identified the binding interface for PDE5 on RhoBTB1 by truncating full-length RhoBTB1 into its component domains. Co-immunoprecipitation analyses revealed that the C-terminal half of RhoBTB1 containing its two BTB domains and the C-terminal domain (B1B2C) is the minimal region required for PDE5 recruitment and subsequent proteasomal degradation via Cullin-3 (CUL3). The C-terminal domain was essential in recruiting PDE5 as constructs lacking this region could not participate in PDE5 binding or proteasomal degradation. We also identified Pro353 and Ser363 as key amino acid residues in the B1B2C region involved in CUL3 binding to RhoBTB1. Mutation of either of these residues exhibited impaired CUL3 binding and PDE5 degradation, although the binding to PDE5 was preserved. Finally, we employed ascorbate peroxidase 2 (APEX2) proximity labeling using a B1B2C-APEX2 fusion protein as bait to capture unknown RhoBTB1 binding partners. Among several B1B2C-binding proteins identified and validated, we focused on SET domain containing 2 (SETD2). SETD2 and RhoBTB1 directly interacted, and the level of SETD2 increased in response to pharmacological inhibition of the proteasome or Cullin complex, CUL3 deletion, and RhoBTB1-inhibition with siRNA. This suggests that SETD2 is regulated by the RhoBTB1-CUL3 axis. Future studies will determine whether SETD2 plays a role in cardiovascular function.
Collapse
Affiliation(s)
- Gaurav Kumar
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shi Fang
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daria Golosova
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ko-Ting Lu
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel T Brozoski
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ibrahim Vazirabad
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Curt D Sigmund
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
7
|
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
|
8
|
Podieh F, Wensveen R, Overboom M, Abbas L, Majolée J, Hordijk P. Differential role for rapid proteostasis in Rho GTPase-mediated control of quiescent endothelial integrity. J Biol Chem 2023; 299:104593. [PMID: 36894017 PMCID: PMC10124901 DOI: 10.1016/j.jbc.2023.104593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Endothelial monolayer permeability is regulated by actin dynamics and vesicular traffic. Recently, ubiquitination was also implicated in the integrity of quiescent endothelium, as it differentially controls the localization and stability of adhesion- and signaling proteins. However, the more general effect of fast protein turnover on endothelial integrity is not clear. Here, we found that inhibition of E1 ubiquitin ligases induces a rapid, reversible loss of integrity in quiescent, primary human endothelial monolayers, accompanied by increased F-actin stress fibers and the formation of intercellular gaps. Concomitantly, total protein and activity of the actin-regulating GTPase RhoB, but not its close homologue RhoA, increase ∼10-fold in 5-8 h. We determined that, the depletion of RhoB, but not of RhoA, the inhibition of actin contractility and the inhibition of protein synthesis all significantly rescue the loss of cell-cell contact induced by E1 ligase inhibition. Collectively, our data suggest that in quiescent human endothelial cells, the continuous and fast turnover of short-lived proteins that negatively regulate cell-cell contact, is essential to preserve monolayer integrity.
Collapse
Affiliation(s)
- Fabienne Podieh
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Roos Wensveen
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - MaxC Overboom
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Lotte Abbas
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Jisca Majolée
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands; Developmental Biology and Stem Cell Research, Hubrecht Institute, 3584 CT, Utrecht, The Netherlands
| | - PeterL Hordijk
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Physiology, De Boelelaan 1117, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Guixé‐Muntet S, Biquard L, Szabo G, Dufour J, Tacke F, Francque S, Rautou P, Gracia‐Sancho J. Review article: vascular effects of PPARs in the context of NASH. Aliment Pharmacol Ther 2022; 56:209-223. [PMID: 35661191 PMCID: PMC9328268 DOI: 10.1111/apt.17046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/04/2021] [Accepted: 05/08/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors known to regulate glucose and fatty acid metabolism, inflammation, endothelial function and fibrosis. PPAR isoforms have been extensively studied in metabolic diseases, including type 2 diabetes and cardiovascular diseases. Recent data extend the key role of PPARs to liver diseases coursing with vascular dysfunction, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). AIM This review summarises and discusses the pathobiological role of PPARs in cardiovascular diseases with a special focus on their impact and therapeutic potential in NAFLD and NASH. RESULTS AND CONCLUSIONS PPARs may be attractive for the treatment of NASH due to their liver-specific effects but also because of their efficacy in improving cardiovascular outcomes, which may later impact liver disease. Assessment of cardiovascular disease in the context of NASH trials is, therefore, of the utmost importance, both from a safety and efficacy perspective.
Collapse
Affiliation(s)
- Sergi Guixé‐Muntet
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain
| | - Louise Biquard
- Université de Paris, Inserm, CNRSCentre de recherche sur l'InflammationUMR1149ParisFrance
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Jean‐François Dufour
- Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCharité Universitätsmedizin Berlin, Campus Virchow‐Klinikum (CVK) and Campus Charité Mitte (CCM)BerlinGermany
| | - Sven Francque
- Department of Gastroenterology and HepatologyAntwerp University HospitalAntwerpBelgium,Translational Sciences in Inflammation and ImmunologyInflaMed Centre of Excellence, Laboratory of Experimental Medicine and Paediatrics, Faculty of Medicine and Health Sciences, University of AntwerpAntwerpBelgium
| | - Pierre‐Emmanuel Rautou
- Université de Paris, AP‐HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGESTCentre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE‐LIVER, Centre de recherche sur l'inflammationParisFrance
| | - Jordi Gracia‐Sancho
- Liver Vascular Biology Research GroupIDIBAPS Biomedical Research Institute & CIBEREHDBarcelonaSpain,Department of Visceral Surgery and Medicine & Department for Biomedical ResearchInselspital, University of BernBernSwitzerland
| |
Collapse
|
10
|
Shen J, Li Y, Li M, Li Z, Deng H, Xie X, Liu J. Restoration of Cullin3 gene expression enhances the improved effects of sonic hedgehog signaling activation for hypertension and attenuates the dysfunction of vascular smooth muscle cells. Biomed Eng Online 2022; 21:39. [PMID: 35715796 PMCID: PMC9206298 DOI: 10.1186/s12938-022-01002-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypertension is known as a major factor for global mortality. We aimed to investigate the role of Cullin3 (CUL3) in the regulation of hypertension. MATERIAL AND METHODS Human vascular smooth muscle cells (VSMCs) were treated with Angiotensin II (Ang II) to establish a hypertension in vitro model. Cell viability was detected by a cell counting kit-8 (CCK-8) assay. The content of reactive oxygen species (ROS) was evaluated by kit. Transwell assay and TUNEL staining were, respectively, used to assess cell migration and apoptosis. Additionally, the expression of sonic hedgehog (SHH) signaling-related proteins (SHH, smoothened homolog (Smo) and glioblastoma (Gli)) and CUL3 was tested with western blotting. Following treatment with Cyclopamine (Cycl), an inhibitor of SHH signaling, in Ang II-induced VSMCs, cell viability, migration, apoptosis and ROS content were determined again. Then, VSMCs were transfected with CUL3 plasmid or/and treated with sonic hedgehog signaling agonist (SAG) to explore the impacts on Ang II-induced VSMCs damage. In vivo, a hypertensive mouse model was established. Systolic blood pressure and diastolic blood pressure were determined. The histopathologic changes of abdominal aortic tissues were examined using H&E staining. The expression of SHH, Smo, Gli and CUL3 was tested with western blotting. RESULTS Significantly increased proliferation, migration and apoptosis of VSMCs were observed after Ang II exposure. Moreover, Ang II induced upregulated SHH, Smo and Gli expression, whereas limited increase in CUL3 expression was observed. The content of ROS in Ang II-stimulated VSMCs presented the same results. Following Cycl treatment, the high levels of proliferation and migration in Ang II-treated VSMCs were notably remedied while the apoptosis and ROS concentration were further increased. Moreover, Cycl downregulated SHH, Smo, Gli and CUL3 expression. Above-mentioned changes caused by Ang II were reversed following SAG addition. Indeed, SAG treatment combined with restoration of CUL3 expression inhibited proliferation, migration, apoptosis and ROS level in Ang II-stimulated VSMCs. In vivo, SAG aggravated the histopathological changes of the aorta and with a worse tendency after both SAG intervention and CUL3 silencing. By contrast, SAG treatment and rebound in CUL3 expression alleviated the vascular damage. CONCLUSIONS Collectively, restoration of CUL3 gene expression protected against hypertension through enhancing the effects of SHH activation in inhibition of apoptosis and oxidative stress for hypertension and alleviating the dysfunction of VSMCs.
Collapse
Affiliation(s)
- Jian Shen
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China.
| | - Youqi Li
- Department of Nephrology, Huizhou Municipal Central Hospital, Huizhou, 516001, Guangdong, China
| | - Menghao Li
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China
| | - Zhiming Li
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China
| | - Huantang Deng
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China
| | - Xiongwei Xie
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China
| | - Jinguang Liu
- Department of Cardiology, Huizhou Municipal Central Hospital, 41 Eling North Road, Huizhou, 516001, Guangdong, China
| |
Collapse
|
11
|
Kelch-like protein 3 in human disease and therapy. Mol Biol Rep 2022; 49:9813-9824. [PMID: 35585379 DOI: 10.1007/s11033-022-07487-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
Kelch-like protein 3 (KLHL3) is a substrate adaptor of Cullin3-RING ubiquitin ligase (CRL3), and KLHL3-CUL3 complex plays a vital role in the ubiquitination of specific substrates. Mutations and abnormal post-translational modifications of KLHL3-CUL3 affect substrate ubiquitination and may related to the pathogenesis of Gordon syndrome (GS), Primary Hyperparathyroidism (PHPT), Diabetes Mellitus (DM), Congenital Heart Disease (CHD), Pre-eclampsia (PE) and even cancers. Therefore, it is essential to understand the function and molecular mechanisms of KLHL3-CUL3 for the treatment of related diseases. In this review, we summary the structure and function of KLHL3-CUL3, the effect of KLHL3-CUL3 mutations and aberrant modifications in GS, PHPT, DM, CHD and PE. Moreover, we noted a possible role of KLHL3-CUL3 in carcinogenesis and provided ideas for targeting KLHL3-CUL3 for related disease treatment.
Collapse
|
12
|
Fang S, Wu J, Reho JJ, Lu KT, Brozoski DT, Kumar G, Werthman AM, Silva SD, Muskus Veitia PC, Wackman KK, Mathison AJ, Teng BQ, Lin CW, Quelle FW, Sigmund CD. RhoBTB1 reverses established arterial stiffness in angiotensin-II hypertension by promoting actin depolymerization. JCI Insight 2022; 7:158043. [PMID: 35358093 PMCID: PMC9090250 DOI: 10.1172/jci.insight.158043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Arterial stiffness predicts cardiovascular disease and all-cause mortality, but its treatment remains challenging. Mice treated with angiotensin II (Ang II) develop hypertension, arterial stiffness, vascular dysfunction, and a downregulation of Rho-related BTB domain–containing protein 1 (RhoBTB1) in the vasculature. RhoBTB1 is associated with blood pressure regulation, but its function is poorly understood. We tested the hypothesis that restoring RhoBTB1 can attenuate arterial stiffness, hypertension, and vascular dysfunction in Ang II–treated mice. Genetic complementation of RhoBTB1 in the vasculature was achieved using mice expressing a tamoxifen-inducible, smooth muscle–specific RhoBTB1 transgene. RhoBTB1 restoration efficiently and rapidly alleviated arterial stiffness but not hypertension or vascular dysfunction. Mechanistic studies revealed that RhoBTB1 had no substantial effect on several classical arterial stiffness contributors, such as collagen deposition, elastin content, and vascular smooth muscle remodeling. Instead, Ang II increased actin polymerization in the aorta, which was reversed by RhoBTB1. Changes in the levels of 2 regulators of actin polymerization, cofilin and vasodilator-stimulated phosphoprotein, in response to RhoBTB1 were consistent with an actin depolymerization mechanism. Our study reveals an important function of RhoBTB1, demonstrates its vital role in antagonizing established arterial stiffness, and further supports a functional and mechanistic separation among hypertension, vascular dysfunction, and arterial stiffness.
Collapse
Affiliation(s)
- Shi Fang
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Jing Wu
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - John J Reho
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Ko-Ting Lu
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Daniel T Brozoski
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Gaurav Kumar
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Alec M Werthman
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Sebastiao Donato Silva
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Patricia C Muskus Veitia
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Kelsey K Wackman
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| | - Angela J Mathison
- Department of Surgery and the Genomic Sciences and Precision Medicine Cente, Medical College of Wisconsin, Milwawkee, United States of America
| | - Bi Qing Teng
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, United States of America
| | - Chien-Wei Lin
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, United States of America
| | - Frederick W Quelle
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, United States of America
| | - Curt D Sigmund
- Department of Physiology and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, United States of America
| |
Collapse
|
13
|
Ertuglu LA, Elijovich F, Laffer CL, Kirabo A. Salt-Sensitivity of Blood Pressure and Insulin Resistance. Front Physiol 2021; 12:793924. [PMID: 34966295 PMCID: PMC8711096 DOI: 10.3389/fphys.2021.793924] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/18/2021] [Indexed: 12/20/2022] Open
Abstract
Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality that is seen in both hypertensive and normotensive populations. Insulin resistance (IR) strongly correlates with SSBP and affects nearly 50% of salt sensitive people. While the precise mechanism by which IR and SSBP relate remains elusive, several common pathways are involved in the genesis of both processes, including vascular dysfunction and immune activation. Vascular dysfunction associated with insulin resistance is characterized by loss of nitric oxide (NO)-mediated vasodilation and heightened endothelin-1 induced vasoconstriction, as well as capillary rarefaction. It manifests with increased blood pressure (BP) in salt sensitive murine models. Another common denominator in the pathogenesis of insulin resistance, hypertension, and salt sensitivity (SS) is immune activation involving pro-inflammatory cytokines like tumor necrosis factor (TNF)-α, IL-1β, and IL-6. In the last decade, a new understanding of interstitial sodium storage in tissues such as skin and muscle has revolutionized traditional concepts of body sodium handling and pathogenesis of SS. We have shown that interstitial Na+ can trigger a T cell mediated inflammatory response through formation of isolevuglandin protein adducts in antigen presenting cells (APCs), and that this response is implicated in salt sensitive hypertension. The peroxisome proliferator-activated receptor γ (PPARγ) is a transcription factor that modulates both insulin sensitivity and BP. PPARγ agonists increase insulin sensitivity and ameliorate salt sensitivity, whereas deficiency of PPARγ results in severe insulin resistance and hypertension. These findings suggest that PPARγ plays a role in the common pathogenesis of insulin sensitivity and salt sensitivity, perhaps via effects on the immune system and vascular function. The goal of this review is to discuss those mechanisms that may play a role in both SSBP and in insulin resistance.
Collapse
Affiliation(s)
- Lale A Ertuglu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
14
|
Hureaux M, Mazurkiewicz S, Boccio V, Vargas-Poussou R, Jeunemaitre X. The variety of genetic defects explains the phenotypic heterogeneity of Familial Hyperkalemic Hypertension. Kidney Int Rep 2021; 6:2639-2652. [PMID: 34622103 PMCID: PMC8484123 DOI: 10.1016/j.ekir.2021.07.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction Familial hyperkalemic hypertension is a rare inherited form of arterial hypertension. Four genes are responsible for this disease, the variants of these genes cause disruption in the regulation of ion transport in the distal renal tubule. Whether the genotype explains the large phenotypic heterogeneity has not been fully explored. Methods We retrospectively analyzed clinical and genetic data of 153 cases (84 probands, 69 relatives) with familial hyperkalemic hypertension. Results Pathogenic variants (25 novel variants) were identified as follows: KLHL3 (n = 50), CUL3 (n = 16), WNK1 acidic motif (n = 11), WNK4 acidic motif (n = 4) and WNK1 intron 1 deletions (n = 3). De novo cases were mainly observed in the CUL3-related cases (9 of 12) and recessive cases were only observed in KLHL3-related cases (14 of 50). More severe forms were observed in recessive KLHL3 and CUL3 cases that were also associated with growth retardation. Patients with WNK1 acidic motif variants had a typical biological phenotype and lower frequency of hypertension conversely to WNK4 variants affecting the same motif. Patients with heterozygous KLHL3 and WNK1 deletions had milder forms. Familial screening in 178 relatives allowed detection and care for 69 positive cases. Blood pressure and hyperkalemia were improved by hydrochlorothiazide in all groups. Conclusions This study confirms the phenotypic variability ranging from the severe and early forms associated with CUL3 and recessive KLHL3 genotypes through intermediate forms associated with KLHL3 dominant, WNK4 and WNK1 deletion to mild form associated with WNK1 acidic motif genotype and reinforces the interest of genetic screening to better orientate medical care and genetic counseling.
Collapse
Affiliation(s)
- Marguerite Hureaux
- Université de Paris, INSERM, Paris Cardiovascular Research Centre, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique et Centre de Référence des Maladies Rénales Héréditaires Rares (MARHEA), Paris, France
| | | | - Valerie Boccio
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique et Centre de Référence des Maladies Rénales Héréditaires Rares (MARHEA), Paris, France
| | - Rosa Vargas-Poussou
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique et Centre de Référence des Maladies Rénales Héréditaires Rares (MARHEA), Paris, France
| | - Xavier Jeunemaitre
- Université de Paris, INSERM, Paris Cardiovascular Research Centre, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique et Centre de Référence des Maladies Rénales Héréditaires Rares (MARHEA), Paris, France
| |
Collapse
|
15
|
Vinton J, Aninweze A, Birgbauer E. Ibuprofen does not inhibit RhoA-mediated growth cone collapse of embryonic chicken retinal axons by LPA. Exp Brain Res 2021; 239:2969-2977. [PMID: 34322723 DOI: 10.1007/s00221-021-06172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/15/2021] [Indexed: 11/28/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that causes neuronal growth cones to collapse and neurites to retract through a RhoA-ROCK mediated pathway. It has been reported that the NSAID ibuprofen improves regeneration after spinal cord injury through a mechanism of inhibiting RhoA. This leads to the hypothesis that ibuprofen should block LPA-mediated growth cone collapse. We tested this hypothesis by treating embryonic chick retinal neurons with ibuprofen followed by LPA. Retinal growth cones collapsed with LPA in the presence of ibuprofen similar to control; however, growth cone collapse was effectively blocked by a ROCK inhibitor. Thus, our results do not support the designation of ibuprofen as a direct RhoA inhibitor.
Collapse
Affiliation(s)
- James Vinton
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Adaeze Aninweze
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA.
| |
Collapse
|
16
|
Podieh F, Hordijk PL. Regulation of Rho GTPases in the Vasculature by Cullin3-Based E3 Ligase Complexes. Front Cell Dev Biol 2021; 9:680901. [PMID: 34136490 PMCID: PMC8201781 DOI: 10.3389/fcell.2021.680901] [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: 03/15/2021] [Accepted: 05/04/2021] [Indexed: 02/02/2023] Open
Abstract
Cullin3-based ubiquitin E3 ligases induce ubiquitination of substrates leading to their proteasomal or lysosomal degradation. BTB proteins serve as adaptors by binding to Cullin3 and recruiting substrate proteins, which enables specific recognition of a broad spectrum of targets. Hence, Cullin3 and its adaptors are involved in myriad cellular processes and organ functions. Cullin3-based ubiquitin E3 ligase complexes target small GTPases of the Rho subfamily, which are key regulators of cytoskeletal dynamics and cell adhesion. In this mini review, we discuss recent insights in Cullin3-mediated regulation of Rho GTPases and their impact on cellular function and disease. Intriguingly, upstream regulators of Rho GTPases are targeted by Cullin3 complexes as well. Thus, Rho GTPase signaling is regulated by Cullin3 on multiple levels. In addition, we address current knowledge of Cullin3 in regulating vascular function, focusing on its prominent role in endothelial barrier function, angiogenesis and the regulation of blood pressure.
Collapse
Affiliation(s)
- Fabienne Podieh
- Department of Physiology, Amsterdam UMC, Amsterdam, Netherlands
| | - Peter L Hordijk
- Department of Physiology, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
17
|
Yao D, He Q, Sun J, Cai L, Wei J, Cai G, Liu J, Lin Y, Wang L, Huang X. FGF21 attenuates hypoxia‑induced dysfunction and inflammation in HPAECs via the microRNA‑27b‑mediated PPARγ pathway. Int J Mol Med 2021; 47:116. [PMID: 33907846 PMCID: PMC8083827 DOI: 10.3892/ijmm.2021.4949] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH), is a chronic and progressive disorder characterized by pulmonary vascular remodeling, including endothelial cell dysfunction and inflammation. MicroRNAs (miRNAs or miRs) play an important role in the development of PAH. In addition, fibroblast growth factor 21 (FGF21) has been found to have marked anti-dysfunction and anti-inflammatory properties. Therefore, the present study aimed to investigate the latent effects of FGF21 against PAH through the miR-27b/peroxisome proliferator-activated receptor γ (PPARγ) axis. Human pulmonary arterial endothelial cells (HPAECs) subjected to hypoxia were used as PAH models. The results revealed that PPARγ expression was downregulated and miR-27b expression was upregulated in the HPAECs exposed to hypoxia. Luciferase assay suggested that PPARγ was a target gene of miR-27b. Furthermore, miR-27b inhibited the expression of the PPARγ gene, thereby aggravating hypoxia-induced HPAEC dysfunction. Moreover, miR-27b activated the nuclear factor-κB signaling pathway and the expression of inflammatory factors [interleukin (IL)-1β, IL-6 and tumor necrosis factor-α] by targeting PPARγ. In addition, the expression of miR-27b decreased following treatment of the hypoxia-exposed HPAECs with FGF21. Furthermore, FGF21 alleviated hypoxia-induced HPAEC dysfunction and inflammation by inhibiting miR-27b expression and thereby promoting PPARγ expression. On the whole, the findings of the present study suggest that FGF21 may serve as a therapeutic target for managing PAH through the miR-27b-mediated PPARγ pathway.
Collapse
Affiliation(s)
- Dan Yao
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Qinlian He
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Junwei Sun
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Luqiong Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Jinqiu Wei
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Gexiang Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Jingjing Liu
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Yinuo Lin
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Liangxing Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
18
|
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
|
19
|
Brown A, Meor Azlan NF, Wu Z, Zhang J. WNK-SPAK/OSR1-NCC kinase signaling pathway as a novel target for the treatment of salt-sensitive hypertension. Acta Pharmacol Sin 2021; 42:508-517. [PMID: 32724175 PMCID: PMC8115323 DOI: 10.1038/s41401-020-0474-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/06/2020] [Indexed: 02/08/2023]
Abstract
Hypertension is the most prevalent health condition worldwide, affecting ~1 billion people. Gordon's syndrome is a form of secondary hypertension that can arise due to a number of possible mutations in key genes that encode proteins in a pathway containing the With No Lysine [K] (WNK) and its downstream target kinases, SPS/Ste20-related proline-alanine-rich kinase (SPAK) and oxidative stress responsive kinase 1 (OSR1). This pathway regulates the activity of the thiazide-sensitive sodium chloride cotransporter (NCC), which is responsible for NaCl reabsorption in the distal nephron. Therefore, mutations in genes encoding proteins that regulate the NCC proteins disrupt ion homeostasis and cause hypertension by increasing NaCl reabsorption. Thiazide diuretics are currently the main treatment option for Gordon's syndrome. However, they have a number of side effects, and chronic usage can lead to compensatory adaptations in the nephron that counteract their action. Therefore, recent research has focused on developing novel inhibitory molecules that inhibit components of the WNK-SPAK/OSR1-NCC pathway, thereby reducing NaCl reabsorption and restoring normal blood pressure. In this review we provide an overview of the currently reported molecular inhibitors of the WNK-SPAK/OSR1-NCC pathway and discuss their potential as treatment options for Gordon's syndrome.
Collapse
Affiliation(s)
- Archie Brown
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
| | - Zhijuan Wu
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK
- Newcastle University Business School, Newcastle University, Newcastle upon Tyne, NE1 4SE, UK
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, Medical School, College of Medicine and Health, University of Exeter, Hatherly Laboratories, Exeter, EX4 4PS, UK.
- Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Xiamen, 361004, China.
| |
Collapse
|
20
|
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: 30] [Impact Index Per Article: 7.5] [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
|
21
|
Wu J, Fang S, Lu KT, Wackman K, Schwartzman ML, Dikalov SI, Grobe JL, Sigmund CD. EP3 (E-Prostanoid 3) Receptor Mediates Impaired Vasodilation in a Mouse Model of Salt-Sensitive Hypertension. Hypertension 2021; 77:1399-1411. [PMID: 33641369 DOI: 10.1161/hypertensionaha.120.16518] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jing Wu
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.)
| | - Shi Fang
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.).,Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa (S.F.)
| | - Ko-Ting Lu
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.)
| | - Kelsey Wackman
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.)
| | - Michal L Schwartzman
- Department of Pharmacology, New York Medical College School of Medicine, Valhalla (M.L.S.)
| | - Sergey I Dikalov
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (S.D.)
| | - Justin L Grobe
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.)
| | - Curt D Sigmund
- From the Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee (J.W., S.F., K.-T.L., K.W., J.L.G., C.D.S.)
| |
Collapse
|
22
|
Wu J, Agbor LN, Fang S, Mukohda M, Nair AR, Nakagawa P, Sharma A, Morgan DA, Grobe JL, Rahmouni K, Weiss RM, McCormick JA, Sigmund CD. Failure to vasodilate in response to salt loading blunts renal blood flow and causes salt-sensitive hypertension. Cardiovasc Res 2021; 117:308-319. [PMID: 32428209 PMCID: PMC7797211 DOI: 10.1093/cvr/cvaa147] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/22/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023] Open
Abstract
AIMS Salt-sensitive (SS) hypertension is accompanied by impaired vasodilation in the systemic and renal circulation. However, the causal relationship between vascular dysfunction and salt-induced hypertension remains controversial. We sought to determine whether primary vascular dysfunction, characterized by a failure to vasodilate during salt loading, plays a causal role in the pathogenesis of SS hypertension. METHODS AND RESULTS Mice selectively expressing a peroxisome proliferator-activated receptor γ dominant-negative mutation in vascular smooth muscle (S-P467L) exhibited progressive SS hypertension during a 4 week high salt diet (HSD). This was associated with severely impaired vasodilation in systemic and renal vessels. Salt-induced impairment of vasodilation occurred as early as 3 days after HSD, which preceded the onset of SS hypertension. Notably, the overt salt-induced hypertension in S-P467L mice was not driven by higher cardiac output, implying elevations in peripheral vascular resistance. In keeping with this, HSD-fed S-P467L mice exhibited decreased smooth muscle responsiveness to nitric oxide (NO) in systemic vessels. HSD-fed S-P467L mice also exhibited elevated albuminuria and a blunted increase in urinary NO metabolites which was associated with blunted renal blood flow and increased sodium retention mediated by a lack of HSD-induced suppression of NKCC2. Blocking NKCC2 function prevented the salt-induced increase in blood pressure in S-P467L mice. CONCLUSION We conclude that failure to vasodilate in response to salt loading causes SS hypertension by restricting renal perfusion and reducing renal NO through a mechanism involving NKCC2 in a mouse model of vascular peroxisome proliferator-activated receptor γ impairment.
Collapse
Affiliation(s)
- Jing Wu
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Larry N Agbor
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Shi Fang
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Masashi Mukohda
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Anand R Nair
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Pablo Nakagawa
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Avika Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR 97239, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Justin L Grobe
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
- Veteran Affairs Health Care System, 601 Hwy 6 West, Iowa City, IA 52242, USA
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - Robert M Weiss
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| | - James A McCormick
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, L334, Portland, OR 97239, USA
| | - Curt D Sigmund
- Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 51 Newton Rd., 2-248 BSB, Iowa City, IA 52242, USA
| |
Collapse
|
23
|
Abstract
This review highlights molecular mechanisms of anti-inflammatory and protective
effects of the nuclear transcription factor, peroxisome proliferator-activated
receptor γ (PPARγ) in vascular tissue. PPARγ is an ubiquitously expressed
nuclear factor, and well-studied in adipose tissue and inflammatory cells.
Additionally, beneficial effects of vascular PPARγ’s on atherosclerosis and
vascular remodeling/dysfunction have been reported although the detailed
mechanism remains to be completely elucidated. Clinical and basic studies have
shown that the synthetic PPARγ ligands, thiazolidinediones (TZDs), have
protective effects against cardiovascular diseases such as atherosclerosis.
Recent studies utilizing genetic tools suggested that those protective effects
of TZDs on cardiovascular diseases are not due to a consequence of improvement
of insulin resistance, but may be due to a direct effect on PPARγ’s in vascular
endothelial and smooth muscle cells. In this review, we discuss proposed
mechanisms by which the vascular PPARγ regulates vascular inflammation and
remodeling/dysfunction especially in smooth muscle cells.
Collapse
Affiliation(s)
- Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Hiroshi Ozaki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| |
Collapse
|
24
|
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
|
25
|
Yamazaki O, Hirohama D, Ishizawa K, Shibata S. Role of the Ubiquitin Proteasome System in the Regulation of Blood Pressure: A Review. Int J Mol Sci 2020; 21:E5358. [PMID: 32731518 PMCID: PMC7432568 DOI: 10.3390/ijms21155358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
The kidney and the vasculature play crucial roles in regulating blood pressure. The ubiquitin proteasome system (UPS), a multienzyme process mediating covalent conjugation of the 76-amino acid polypeptide ubiquitin to a substrate protein followed by proteasomal degradation, is involved in multiple cellular processes by regulating protein turnover in various tissues. Increasing evidence demonstrates the roles of UPS in blood pressure regulation. In the kidney, filtered sodium is reabsorbed through diverse sodium transporters and channels along renal tubules, and studies conducted till date have provided insights into the complex molecular network through which ubiquitin ligases modulate sodium transport in different segments. Components of these pathways include ubiquitin ligase neuronal precursor cell-expressed developmentally downregulated 4-2, Cullin-3, and Kelch-like 3. Moreover, accumulating data indicate the roles of UPS in blood vessels, where it modulates nitric oxide bioavailability and vasoconstriction. Cullin-3 not only regulates renal salt reabsorption but also controls vascular tone using different adaptor proteins that target distinct substrates in vascular smooth muscle cells. In endothelial cells, UPS can also contribute to blood pressure regulation by modulating endothelial nitric oxide synthase. In this review, we summarize current knowledge regarding the role of UPS in blood pressure regulation, focusing on renal sodium reabsorption and vascular function.
Collapse
Affiliation(s)
| | | | | | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8605, Japan; (O.Y.); (D.H.); (K.I.)
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW This review provides an up-to-date understanding of how peroxisome proliferator activated receptor γ (PPARγ) exerts its cardioprotective effect in the vasculature through its activation of novel PPARγ target genes in endothelium and vascular smooth muscle. RECENT FINDINGS In vascular endothelial cells, PPARγ plays a protective role by increasing nitric oxide bioavailability and preventing oxidative stress. RBP7 is a PPARγ target gene enriched in vascular endothelial cells, which is likely to form a positive feedback loop with PPARγ. In vascular smooth muscle cells, PPARγ antagonizes the renin-angiotensin system, maintains vascular integrity, suppresses vasoconstriction, and promotes vasodilation through distinct pathways. Rho-related BTB domain containing protein 1 (RhoBTB1) is a novel PPARγ gene target in vascular smooth muscle cells that mediates the protective effect of PPARγ by serving as a substrate adaptor between the Cullin-3 RING ubiquitin ligase and phosphodiesterase 5, thus restraining its activity through ubiquitination and proteasomal degradation. SUMMARY In the vasculature, PPARγ exerts its cardioprotective effect through its transcriptional activity in endothelium and vascular smooth muscle. From the understanding of PPARγ's transcription targets in those pathways, novel hypertension therapy target(s) will emerge.
Collapse
|
27
|
Rafael C, Hadchouel J. [Severity of familial hyperkalemic hypertension caused by CUL-3 mutations: a story about kidneys and blood vessels]. Med Sci (Paris) 2020; 36:455-458. [PMID: 32452365 DOI: 10.1051/medsci/2020074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Chloé Rafael
- Centre de recherche des Cordeliers, Inserm UMR-S1138, Sorbonne université, Université de Paris, 15 rue de l'École de Médecine, 75006 Paris, France. - CNRS ERL 8228 - Laboratoire de physiologie rénale et tubulopathies, 75006, Paris, France
| | - Juliette Hadchouel
- Inserm UMR_S1155, Hôpital Tenon, 4 rue de la Chine, 75020 Paris, France. - Faculté de médecine, Sorbonne université, Paris, France
| |
Collapse
|
28
|
Sigmund CD, Carey RM, Appel L, Arnett D, Bosworth HB, Cushman WC, Galis ZS, Parker MG, Hall JE, Harrison DG, McDonough AA, Nicastro HL, Oparil S, Osborn JW, Raizada MK, Wright JD, Oh YS. Report of the National Heart, Lung, and Blood Institute Working Group on Hypertension: Barriers to Translation. Hypertension 2020; 75:902-917. [PMID: 32063061 PMCID: PMC7067675 DOI: 10.1161/hypertensionaha.119.13887] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The National Heart, Lung, and Blood Institute convened a multidisciplinary working group of hypertension researchers on December 6 to 7, 2018, in Bethesda, MD, to share current scientific knowledge in hypertension and to identify barriers to translation of basic into clinical science/trials and implementation of clinical science into clinical care of patients with hypertension. The goals of the working group were (1) to provide an overview of recent discoveries that may be ready for testing in preclinical and clinical studies; (2) to identify gaps in knowledge that impede translation; (3) to highlight the most promising scientific areas in which to pursue translation; (4) to identify key challenges and barriers for moving basic science discoveries into translation, clinical studies, and trials; and (5) to identify roadblocks for effective dissemination and implementation of basic and clinical science in real-world settings. The working group addressed issues that were responsive to many of the objectives of the National Heart, Lung, and Blood Institute Strategic Vision. The working group identified major barriers and opportunities for translating research to improved control of hypertension. This review summarizes the discussion and recommendations of the working group.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - John E. Hall
- University of Mississippi Medical Center, Jackson, MS
| | | | | | | | | | | | | | | | - Young S. Oh
- Vascular Biology & Hypertension Branch, DCVS, NHLBI
| |
Collapse
|
29
|
Wu L, Guo C, Wu J. Therapeutic potential of PPARγ natural agonists in liver diseases. J Cell Mol Med 2020; 24:2736-2748. [PMID: 32031298 PMCID: PMC7077554 DOI: 10.1111/jcmm.15028] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 11/17/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator‐activated receptor gamma (PPARγ) is a vital subtype of the PPAR family. The biological functions are complex and diverse. PPARγ plays a significant role in protecting the liver from inflammation, oxidation, fibrosis, fatty liver and tumours. Natural products are a promising pool for drug discovery, and enormous research effort has been invested in exploring the PPARγ‐activating potential of natural products. In this manuscript, we will review the research progress of PPARγ agonists from natural products in recent years and probe into the application potential and prospects of PPARγ natural agonists in the therapy of various liver diseases, including inflammation, hepatic fibrosis, non‐alcoholic fatty liver and liver cancer.
Collapse
Affiliation(s)
- Liwei Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chuanyong Guo
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianye Wu
- Department of Gastroenterology, Putuo People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Abstract
Essential hypertension is a highly prevalent disease in the general population. Secondary hypertension is characterized by a specific and potentially reversible cause of increased blood pressure levels. Some secondary endocrine forms of hypertension are common (caused by uncontrolled cortisol, aldosterone, or catecholamines production). This article describes rare monogenic forms of hypertension, characterized by electrolyte disorders and suppressed renin-aldosterone axis. They represent simple models for the physiology of renal control of sodium levels and plasma volume, thus reaching a high scientific interest. Furthermore, they could explain some features closer to the essential phenotype of hypertension, suggesting a mechanistically driven personalized treatment.
Collapse
MESH Headings
- Adrenal Hyperplasia, Congenital/complications
- Adrenal Hyperplasia, Congenital/metabolism
- Adrenal Hyperplasia, Congenital/therapy
- Arthrogryposis/complications
- Arthrogryposis/metabolism
- Arthrogryposis/therapy
- Cleft Palate/complications
- Cleft Palate/metabolism
- Cleft Palate/therapy
- Clubfoot/complications
- Clubfoot/metabolism
- Clubfoot/therapy
- Hand Deformities, Congenital/complications
- Hand Deformities, Congenital/metabolism
- Hand Deformities, Congenital/therapy
- Humans
- Hypertension/drug therapy
- Hypertension/etiology
- Hypertension/metabolism
- Hypertension/physiopathology
- Liddle Syndrome/complications
- Liddle Syndrome/metabolism
- Liddle Syndrome/therapy
- Mineralocorticoid Excess Syndrome, Apparent/complications
- Mineralocorticoid Excess Syndrome, Apparent/metabolism
- Mineralocorticoid Excess Syndrome, Apparent/therapy
- Mineralocorticoid Excess Syndrome, Apparent
Collapse
Affiliation(s)
- Filippo Ceccato
- Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padova, Via Ospedale Civile, 105, Padova 35128, Italy.
| | - Franco Mantero
- Endocrinology Unit, Department of Medicine DIMED, University-Hospital of Padova, Via Ospedale Civile, 105, Padova 35128, Italy
| |
Collapse
|
31
|
Mukohda M. [Role of PPARγ, a transcription factor in cardiovascular disease]. Nihon Yakurigaku Zasshi 2019; 154:56-60. [PMID: 31406043 DOI: 10.1254/fpj.154.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand activated transcription factor known to regulate fatty acid metabolism. Thiazolidinediones (TZDs), PPARγ synthetic agonists, currently used to treat patients with type 2 diabetes, have been shown to lower the blood pressure and protect against vascular diseases such as atherosclerosis. In line with these findings, it has been reported that individuals with loss-of-function mutations of PPARγ developed sever early-onset hypertension in addition to metabolic abnormalities. Accumulating evidences suggest PPARγ in the vasculature has protective effects on cardiovascular disease despite unclear mechanism. Because of ubiquitous expression of PPARγ, TZDs are well-known to be associated with serious side effects such as weight gain, fluid retention, and bone fractures. Thus identification of mechanisms on tissue-specific PPARγ activity may lead to the development of targeted treatment which is characterized by no deleterious effects. This review discusses role of PPARγ in cardiovascular disease.
Collapse
Affiliation(s)
- Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Mukohda M, Fang S, Wu J, Agbor LN, Nair AR, Ibeawuchi SRC, Hu C, Liu X, Lu KT, Guo DF, Davis DR, Keen HL, Quelle FW, Sigmund CD. RhoBTB1 protects against hypertension and arterial stiffness by restraining phosphodiesterase 5 activity. J Clin Invest 2019; 129:2318-2332. [PMID: 30896450 DOI: 10.1172/jci123462] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.
Collapse
Affiliation(s)
- Masashi Mukohda
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Shi Fang
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jing Wu
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Larry N Agbor
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Anand R Nair
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Stella-Rita C Ibeawuchi
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Chunyan Hu
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Xuebo Liu
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Ko-Ting Lu
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Deng-Fu Guo
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Deborah R Davis
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Henry L Keen
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Frederick W Quelle
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Curt D Sigmund
- Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Department of Physiology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
35
|
Chen M, Shen H, Zhu L, Yang H, Ye P, Liu P, Gu Y, Chen S. Berberine attenuates hypoxia-induced pulmonary arterial hypertension via bone morphogenetic protein and transforming growth factor-β signaling. J Cell Physiol 2019; 234:17482-17493. [PMID: 30786011 DOI: 10.1002/jcp.28370] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/30/2022]
Abstract
Hypoxia-induced excessive pulmonary artery smooth muscle cell (PASMC) proliferation plays an important role in the pathology of pulmonary arterial hypertension (PAH). Berberine (BBR) is reported as an effective antiproliferative properties applied in clinical. However, the effect of BBR on PAH remains unclear. In the present study, we elucidated the protective effects of BBR against abnormal PASMC proliferation and vascular remodeling in chronic hypoxia-induced hearts. Furthermore, the potential mechanisms of BBR were investigated. For this purpose, C57/BL6 mice were exposed to chronic hypoxia for 4 weeks to mimic severe PAH. Hemodynamic and pulmonary pathomorphology data showed that chronic hypoxia significantly increased the right ventricular systolic pressure (RVSP), the right ventricle/left ventricle plus septum RV/(LV + S) weight ratio, and the median width of pulmonary arterioles. BBR attenuated the elevations in RVSP and RV/(LV + S) and mitigated pulmonary vascular structure remodeling. BBR also suppressed the hypoxia-induced increases in the expression of proliferating cell nuclear antigen (PCNA) and of α-smooth muscle actin. Furthermore, administration of BBR significantly increased the expression of bone morphogenetic protein type II receptor (BMPR-II) and its downstream molecules P-smad1/5 and decreased the expression of transforming growth factor-β (TGF-β) and its downstream molecules P-smad2/3. Moreover, peroxisome proliferator-activated receptor γ expression was significantly decreased in the hypoxia group, and this decrease was reversed by BBR treatment. Our study demonstrated that the protective effect of BBR against hypoxia-induced PAH in a mouse model may be achieved through altered BMPR-II and TGF-β signaling.
Collapse
Affiliation(s)
- Mingxing Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hui Shen
- Department of Cardiology, The affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Linlin Zhu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hongfeng Yang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Peng Ye
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Pengfei Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shaoliang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
36
|
Dee RA, Mangum KD, Bai X, Mack CP, Taylor JM. Druggable targets in the Rho pathway and their promise for therapeutic control of blood pressure. Pharmacol Ther 2019; 193:121-134. [PMID: 30189292 PMCID: PMC7235948 DOI: 10.1016/j.pharmthera.2018.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The prevalence of high blood pressure (also known as hypertension) has steadily increased over the last few decades. Known as a silent killer, hypertension increases the risk for cardiovascular disease and can lead to stroke, heart attack, kidney failure and associated sequela. While numerous hypertensive therapies are currently available, it is estimated that only half of medicated patients exhibit blood pressure control. This signifies the need for a better understanding of the underlying cause of disease and for more effective therapies. While blood pressure homeostasis is very complex and involves the integrated control of multiple body systems, smooth muscle contractility and arterial resistance are important contributors. Strong evidence from pre-clinical animal models and genome-wide association studies indicate that smooth muscle contraction and BP homeostasis are governed by the small GTPase RhoA and its downstream target, Rho kinase. In this review, we summarize the signaling pathways and regulators that impart tight spatial-temporal control of RhoA activity in smooth muscle cells and discuss current therapeutic strategies to target these RhoA pathway components. We also discuss known allelic variations in the RhoA pathway and consider how these polymorphisms may affect genetic risk for hypertension and its clinical manifestations.
Collapse
Affiliation(s)
- Rachel A Dee
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kevin D Mangum
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Xue Bai
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Christopher P Mack
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Joan M Taylor
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
37
|
Peroxisome proliferator-activated receptor-gamma activation attenuates diabetic cardiomyopathy via regulation of the TGF-β/ERK pathway and epithelial-to-mesenchymal transition. Life Sci 2018; 213:269-278. [DOI: 10.1016/j.lfs.2018.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/11/2018] [Accepted: 09/01/2018] [Indexed: 12/17/2022]
|
38
|
Ma TJ, Zhang ZW, Lu YL, Zhang YY, Tao DC, Liu YQ, Ma YX. CLOCK and BMAL1 stabilize and activate RHOA to promote F-actin formation in cancer cells. Exp Mol Med 2018; 50:1-15. [PMID: 30287810 PMCID: PMC6172197 DOI: 10.1038/s12276-018-0156-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 02/05/2023] Open
Abstract
Circadian genes control most of the physiological functions in cancer cells, including cell proliferation, migration, and invasion. The CLOCK and BMAL1 complex plays a central role in circadian rhythms. Previous studies have shown that circadian genes may act as oncogenes or tumor-suppressor genes. In addition, F-actin, regulated by RHOA, has been shown to participate in tumor progression. However, the roles of the CLOCK and BMAL1 genes in the regulation of tumor progression via the RHOA-ROCK-CFL pathway remain largely unclear. Here we first indicate that the rearrangement of F-actin is regulated by CLOCK and BMAL1. We found that CLOCK and BMAL1 can upregulate RHOA expression by inhibiting CUL3-mediated ubiquitination and activate RHOA by reducing the interaction between RHOA and RhoGDI. Consequently, CLOCK and BMAL1 control the expression of the components of the RHOA-ROCK-CFL pathway, which alters the dynamics of F-actin/G-actin turnover and promotes cancer cell proliferation, migration, and invasion. In conclusion, our research proposes a novel insight into the role of CLOCK and BMAL1 in tumor cells.
Collapse
Affiliation(s)
- Teng-Jiao Ma
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Zhi-Wei Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Yi-Lu Lu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Ying-Ying Zhang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Da-Chang Tao
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Yun-Qiang Liu
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China
| | - Yong-Xin Ma
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital and Collaborative Innovation Center, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
39
|
Raghavan S, Singh NK, Mani AM, Rao GN. Protease-activated receptor 1 inhibits cholesterol efflux and promotes atherogenesis via cullin 3-mediated degradation of the ABCA1 transporter. J Biol Chem 2018; 293:10574-10589. [PMID: 29777060 DOI: 10.1074/jbc.ra118.003491] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 05/10/2018] [Indexed: 12/25/2022] Open
Abstract
Although signaling of thrombin via its receptor protease-activated receptor 1 (Par1) is known to occur in atherothrombosis, its link to the actual pathogenesis of this condition is less clear. To better understand the role of thrombin-Par1 signaling in atherosclerosis, here we have studied their effects on cellular cholesterol efflux in mice. We found that by activating Par1 and cullin 3-mediated ubiquitination and degradation of ABC subfamily A member 1 (ABCA1), thrombin inhibits cholesterol efflux in both murine macrophages and smooth muscle cells. Moreover, disruption of the Par1 gene rescued ABCA1 from Western diet-induced ubiquitination and degradation and restored cholesterol efflux in apolipoprotein E-deficient (ApoE-/-) mice. Similarly, the Par1 deletion diminished diet-induced atherosclerotic lesions in the ApoE-/- mice. These observations for the first time indicate a role for thrombin-Par1 signaling in the pathogenesis of diet-induced atherosclerosis. We identify cullin 3 as a cullin-RING ubiquitin E3 ligase that mediates ABCA1 ubiquitination and degradation and thereby inhibits cholesterol efflux. Furthermore, compared with peripheral blood mononuclear cells (PBMCs) from ApoE-/- mice, the PBMCs from ApoE-/-:Par1-/- mice exhibited decreased trafficking to inflamed arteries of Western diet-fed ApoE-/- mice. This finding suggested that besides inhibiting cholesterol efflux, thrombin-Par1 signaling also plays a role in the recruitment of leukocytes during diet-induced atherogenesis. Based on these findings, we conclude that thrombin-Par1 signaling appears to contribute to the pathogenesis of atherosclerosis by impairing cholesterol efflux from cells and by recruiting leukocytes to arteries.
Collapse
Affiliation(s)
- Somasundaram Raghavan
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Nikhlesh K Singh
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Arul M Mani
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Gadiparthi N Rao
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| |
Collapse
|
40
|
Boerman EM, Sen S, Shaw RL, Joshi T, Segal SS. Gene expression profiles of ion channels and receptors in mouse resistance arteries: Effects of cell type, vascular bed, and age. Microcirculation 2018; 25:e12452. [PMID: 29577514 PMCID: PMC5949082 DOI: 10.1111/micc.12452] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/19/2018] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Receptors and ion channels of smooth muscle cells (SMCs) and endothelial cells (ECs) are integral to the regulation of vessel diameter and tissue blood flow. Physiological roles of ion channels and receptors in skeletal muscle and mesenteric arteries have been identified; however, their gene expression profiles are undefined. We tested the hypothesis that expression profiles for ion channels and receptors governing vascular reactivity vary with cell type, vascular bed, and age. METHODS Mesenteric and superior epigastric arteries were dissected from Old (24-26 months) and Young (3-6 months) C57BL/6J mice. ECs and SMCs were collected for analysis with custom qRT-PCR arrays to determine expression profiles of 80 ion channel and receptor genes. Bioinformatics analyses were applied to gain insight into functional interactions. RESULTS We identified 68 differences in gene expression with respect to cell type, vessel type, and age. Heat maps illustrate differential expression, and distance matrices predict patterns of coexpression. Gene networks based upon protein-protein interaction datasets and KEGG pathways illustrate biological processes affected by specific differences in gene expression. CONCLUSIONS Differences in gene expression profiles are most pronounced between microvascular ECs and SMCs with subtle variations between vascular beds and age groups.
Collapse
Affiliation(s)
- Erika M. Boerman
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Sidharth Sen
- MU Informatics Institute, University of Missouri, Columbia, MO 65211
| | - Rebecca L. Shaw
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212
| | - Trupti Joshi
- MU Informatics Institute, University of Missouri, Columbia, MO 65211
- Health Management and Informatics and Office of Research, School of Medicine, University of Missouri, Columbia, MO 65212
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211
| | - Steven S. Segal
- Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212
- Dalton Cardiovascular Research Center, Columbia, MO 65211
| |
Collapse
|
41
|
Jin J, Jing Z, Ye Z, Guo L, Hua L, Wang Q, Wang J, Cheng Q, Zhang J, Xu Y, Wei L. MLN4924 suppresses lipopolysaccharide-induced proinflammatory cytokine production in neutrophils in a dose-dependent manner. Oncol Lett 2018; 15:8039-8045. [PMID: 29849806 DOI: 10.3892/ol.2018.8333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/01/2017] [Indexed: 12/14/2022] Open
Abstract
Neddylation is a ubiquitination-like pathway. It has been reported that neddylation inhibition with the pharmacological agent MLN4924 potently uppresses lipopolysaccharide (LPS)-induced proinflammatory cytokine production, including tumor necrosis factor (TNF)-α and interleukin (IL)-6, by preventing the degradation of phosphorylated inhibitor of κB (p-IκB) in macrophages. However, whether neddylation serves a similar role in neutrophils remains unknown. In the present study MLN4924 treatment led to the accumulation of P-IκBα in neutrophils as well as the decreased production of TNF-α, IL-6 and IL-1β in response to LPS, in a dose-dependent manner. The viability of neutrophils was only marginally affected in the same conditions, without statistical significance. Furthermore, the nuclear factor (NF)-κB inhibitor JSH-23 mimicked the effects of MLN4924 in neutrophils, and the inhibitory effects of MLN4924 on LPS-induced proinflammatory cytokine production diminished in the presence of JSH-23. Thus, the results of the present study suggest that neddylation inhibition suppresses neutrophil function by suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jiayang Jin
- Laboratory of Immunology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Zhaofei Jing
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China.,Laboratory of Snake Venom, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Zhenjie Ye
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China.,Laboratory of Snake Venom, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Lu Guo
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China.,Henan Key Laboratory of Cellular and Molecular Immunology, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Lei Hua
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China.,College of Pharmacy, Henan University, Kaifeng, Henan 475004, P.R. China
| | - Qingyang Wang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Jing Wang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Qianqian Cheng
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Jiyan Zhang
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Yunlu Xu
- Laboratory of Snake Venom, Fujian Medical University, Fuzhou, Fujian 350004, P.R. China
| | - Lin Wei
- Laboratory of Immunology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
42
|
The NEDD8-activating enzyme inhibitor MLN4924 induces G2 arrest and apoptosis in T-cell acute lymphoblastic leukemia. Oncotarget 2018; 7:23812-24. [PMID: 26993774 PMCID: PMC5029665 DOI: 10.18632/oncotarget.8068] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 02/29/2016] [Indexed: 01/08/2023] Open
Abstract
The first-in-class compound MLN4924 is a small molecule inhibitor that selectively inactivates NEDD8-activating enzyme (NAE). The anticancer effects of MLN4924 have been attributed to impaired neddylation of Cullin proteins. Here, we show that treatment of T-cell acute lymphoblastic leukemia (T-ALL) cells with MLN4924 potently suppressed the neddylation of Cullins and the oncogenic growth of T-ALL cells in-vitro. Moreover, MLN4924 induced disease regression in an in vivo xenograft model. MLN4924 also induced cell cycle arrest at G2 phase and apoptosis in T-ALL cells. However, inhibition of the neddylation of Cullins alone could not explain the effects of MLN4924 in T-ALL cells. Gene expression profiling indicated ribosome function, steroid biosynthesis, and hematopoietic cell lineage pathways were affected by MLN4924 treatment. MLN4924 also induced nucleolar disruption, suggesting nucleolar stress signaling might contribute to the anticancer effects of MLN4924 in T-ALL cells. In addition, MLN4924 treatment reduced 14-3-3ξ\δ protein levels in T-ALL cells. Thus, MLN4924 may inhibit T-ALL cell proliferation via several pathways.
Collapse
|
43
|
Hensley MK, Levine A, Gladwin MT, Lai YC. Emerging therapeutics in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2018; 314:L769-L781. [PMID: 29388467 DOI: 10.1152/ajplung.00259.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pulmonary hypertension (PH) is a progressive and often fatal illness presenting with nonspecific symptoms of dyspnea, lower extremity edema, and exercise intolerance. Pathologically, endothelial dysfunction leads to abnormal intimal and smooth muscle proliferation along with reduced apoptosis, resulting in increased pulmonary vascular resistance and elevated pulmonary pressures. PH is subdivided into five World Health Organization groups based on the disease pathology and specific cause. While there are Food and Drug Administration-approved medications for the treatment of pulmonary arterial hypertension (PAH; Group 1 PH), as well as for chronic thromboembolic PH (Group 4 PH), the morbidity and mortality remain high. Moreover, there are no approved therapies for other forms of PH (Groups 2, 3, and 5) at present. New research has identified molecular targets that mediate vasodilation, anti-inflammatory, and antifibrotic changes within the pulmonary vasculature. Given that PAH is the most commonly studied form of PH worldwide and because recent studies have led to better mechanistic understanding of this devastating disease, in this review we attempt to provide an updated overview of new therapeutic approaches under investigation for the treatment of PH, with a particular focus on PAH, as well as to offer guidelines for future investigations.
Collapse
Affiliation(s)
- Matthew K Hensley
- Division of Pulmonary and Critical Care Medicine, University of Michigan , Ann Arbor, Michigan
| | - Andrea Levine
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Yen-Chun Lai
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
44
|
Functional analysis of Cullin 3 E3 ligases in tumorigenesis. Biochim Biophys Acta Rev Cancer 2017; 1869:11-28. [PMID: 29128526 DOI: 10.1016/j.bbcan.2017.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/14/2022]
Abstract
Cullin 3-RING ligases (CRL3) play pivotal roles in the regulation of various physiological and pathological processes, including neoplastic events. The substrate adaptors of CRL3 typically contain a BTB domain that mediates the interaction between Cullin 3 and target substrates to promote their ubiquitination and subsequent degradation. The biological implications of CRL3 adaptor proteins have been well described where they have been found to play a role as either an oncogene, tumor suppressor, or can mediate either of these effects in a context-dependent manner. Among the extensively studied CRL3-based E3 ligases, the role of the adaptor protein SPOP (speckle type BTB/POZ protein) in tumorigenesis appears to be tissue or cellular context dependent. Specifically, SPOP acts as a tumor suppressor via destabilizing downstream oncoproteins in many malignancies, especially in prostate cancer. However, SPOP has largely an oncogenic role in kidney cancer. Keap1, another well-characterized CRL3 adaptor protein, likely serves as a tumor suppressor within diverse malignancies, mainly due to its specific turnover of its downstream oncogenic substrate, NRF2 (nuclear factor erythroid 2-related factor 2). In accordance with the physiological role the various CRL3 adaptors exhibit, several pharmacological agents have been developed to disrupt its E3 ligase activity, therefore blocking its potential oncogenic activity to mitigate tumorigenesis.
Collapse
|
45
|
Kctd13 deletion reduces synaptic transmission via increased RhoA. Nature 2017; 551:227-231. [PMID: 29088697 DOI: 10.1038/nature24470] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 10/04/2017] [Indexed: 11/08/2022]
Abstract
Copy-number variants of chromosome 16 region 16p11.2 are linked to neuropsychiatric disorders and are among the most prevalent in autism spectrum disorders. Of many 16p11.2 genes, Kctd13 has been implicated as a major driver of neurodevelopmental phenotypes. The function of KCTD13 in the mammalian brain, however, remains unknown. Here we delete the Kctd13 gene in mice and demonstrate reduced synaptic transmission. Reduced synaptic transmission correlates with increased levels of Ras homolog gene family, member A (RhoA), a KCTD13/CUL3 ubiquitin ligase substrate, and is reversed by RhoA inhibition, suggesting increased RhoA as an important mechanism. In contrast to a previous knockdown study, deletion of Kctd13 or kctd13 does not increase brain size or neurogenesis in mice or zebrafish, respectively. These findings implicate Kctd13 in the regulation of neuronal function relevant to neuropsychiatric disorders and clarify the role of Kctd13 in neurogenesis and brain size. Our data also reveal a potential role for RhoA as a therapeutic target in disorders associated with KCTD13 deletion.
Collapse
|
46
|
De Silva TM, Hu C, Kinzenbaw DA, Modrick ML, Sigmund CD, Faraci FM. Genetic Interference With Endothelial PPAR-γ (Peroxisome Proliferator-Activated Receptor-γ) Augments Effects of Angiotensin II While Impairing Responses to Angiotensin 1-7. Hypertension 2017; 70:559-565. [PMID: 28674038 DOI: 10.1161/hypertensionaha.117.09358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 03/27/2017] [Accepted: 05/31/2017] [Indexed: 12/25/2022]
Abstract
Pharmacological activation of PPAR-γ (peroxisome proliferator-activated receptor-γ) protects the vasculature. Much less is known on the cell-specific impact of PPAR-γ when driven by endogenous ligands. Recently, we found that endothelial PPAR-γ protects against angiotensin II-induced endothelial dysfunction. Here, we explored that concept further examining whether effects were sex dependent along with underlying mechanisms. We studied mice expressing a human dominant-negative mutation in PPAR-γ driven by the endothelial-specific vascular cadherin promoter (E-V290M), using nontransgenic littermates as controls. Acetylcholine (an endothelium-dependent agonist) produced similar relaxation of carotid arteries from nontransgenic and E-V290M mice. Incubation of isolated arteries with angiotensin II (1 nmol/L) overnight had no effect in nontransgenic, but reduced responses to acetylcholine by about 50% in male and female E-V290M mice (P<0.05). Endothelial function in E-V290M mice was restored to normal by inhibitors of superoxide (tempol), NADPH oxidase (VAS-2870), Rho kinase (Y-27632), ROCK2 (SLX-2119), NF-κB (nuclear factor-kappa B essential modulator-binding domain peptide), or interleukin-6 (neutralizing antibody). In addition, we hypothesized that PPAR-γ may influence the angiotensin 1-7 arm of the renin-angiotensin system. In the basilar artery, dilation to angiotensin 1-7 was selectively reduced in E-V290M mice by >50% (P<0.05), an effect reversed by Y-27632. Thus, effects of angiotensin II are augmented by interference with endothelial PPAR-γ through sex-independent mechanisms, involving oxidant-inflammatory signaling and ROCK2 (Rho kinase). The study also provides the first evidence that endothelial PPAR-γ interacts with angiotensin 1-7 responses. These critical roles for endothelial PPAR-γ have implications for pathophysiology and therapeutic approaches for vascular disease.
Collapse
Affiliation(s)
- T Michael De Silva
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Chunyan Hu
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Dale A Kinzenbaw
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Mary L Modrick
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Curt D Sigmund
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.)
| | - Frank M Faraci
- From the Departments of Internal Medicine (T.M.D.S., D.A.K., M.L.M., F.M.F.) and Pharmacology (C.H., C.D.S., F.M.F.), Center for Hypertension Research, Carver College of Medicine, The University of Iowa; and Iowa City Veterans Affairs Healthcare System (F.M.F.).
| |
Collapse
|
47
|
Mukohda M, Lu KT, Guo DF, Wu J, Keen HL, Liu X, Ketsawatsomkron P, Stump M, Rahmouni K, Quelle FW, Sigmund CD. Hypertension-Causing Mutation in Peroxisome Proliferator-Activated Receptor γ Impairs Nuclear Export of Nuclear Factor-κB p65 in Vascular Smooth Muscle. Hypertension 2017; 70:174-182. [PMID: 28507170 DOI: 10.1161/hypertensionaha.117.09276] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/08/2017] [Accepted: 04/08/2017] [Indexed: 12/31/2022]
Abstract
Selective expression of dominant negative (DN) peroxisome proliferator-activated receptor γ (PPARγ) in vascular smooth muscle cells (SMC) results in hypertension, atherosclerosis, and increased nuclear factor-κB (NF-κB) target gene expression. Mesenteric SMC were cultured from mice designed to conditionally express wild-type (WT) or DN-PPARγ in response to Cre recombinase to determine how SMC PPARγ regulates expression of NF-κB target inflammatory genes. SMC-specific overexpression of WT-PPARγ or agonist-induced activation of endogenous PPARγ blunted tumor necrosis factor α (TNF-α)-induced NF-κB target gene expression and activity of an NF-κB-responsive promoter. TNF-α-induced gene expression responses were enhanced by DN-PPARγ in SMC. Although expression of NF-κB p65 was unchanged, nuclear export of p65 was accelerated by WT-PPARγ and prevented by DN-PPARγ in SMC. Leptomycin B, a nuclear export inhibitor, blocked p65 nuclear export and inhibited the anti-inflammatory action of PPARγ. Consistent with a role in facilitating p65 nuclear export, WT-PPARγ coimmunoprecipitated with p65, and WT-PPARγ was also exported from the nucleus after TNF-α treatment. Conversely, DN-PPARγ does not bind to p65 and was retained in the nucleus after TNF-α treatment. Transgenic mice expressing WT-PPARγ or DN-PPARγ specifically in SMC (S-WT or S-DN) were bred with mice expressing luciferase controlled by an NF-κB-responsive promoter to assess effects on NF-κB activity in whole tissue. TNF-α-induced NF-κB activity was decreased in aorta and carotid artery from S-WT but was increased in vessels from S-DN mice. We conclude that SMC PPARγ blunts expression of proinflammatory genes by inhibition of NF-κB activity through a mechanism promoting nuclear export of p65, which is abolished by DN mutation in PPARγ.
Collapse
Affiliation(s)
- Masashi Mukohda
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Ko-Ting Lu
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Deng-Fu Guo
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Jing Wu
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Henry L Keen
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Xuebo Liu
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Pimonrat Ketsawatsomkron
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Madeliene Stump
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Kamal Rahmouni
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Frederick W Quelle
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa
| | - Curt D Sigmund
- From the Department of Pharmacology and UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa.
| |
Collapse
|
48
|
Calvier L, Chouvarine P, Legchenko E, Hoffmann N, Geldner J, Borchert P, Jonigk D, Mozes MM, Hansmann G. PPARγ Links BMP2 and TGFβ1 Pathways in Vascular Smooth Muscle Cells, Regulating Cell Proliferation and Glucose Metabolism. Cell Metab 2017; 25:1118-1134.e7. [PMID: 28467929 DOI: 10.1016/j.cmet.2017.03.011] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 12/21/2016] [Accepted: 03/20/2017] [Indexed: 01/24/2023]
Abstract
BMP2 and TGFβ1 are functional antagonists of pathological remodeling in the arteries, heart, and lung; however, the mechanisms in VSMCs, and their disturbance in pulmonary arterial hypertension (PAH), are unclear. We found a pro-proliferative TGFβ1-Stat3-FoxO1 axis in VSMCs, and PPARγ as inhibitory regulator of TGFβ1-Stat3-FoxO1 and TGFβ1-Smad3/4, by physically interacting with Stat3 and Smad3. TGFβ1 induces fibrosis-related genes and miR-130a/301b, suppressing PPARγ. Conversely, PPARγ inhibits TGFβ1-induced mitochondrial activation and VSMC proliferation, and regulates two glucose metabolism-related enzymes, platelet isoform of phosphofructokinase (PFKP, a PPARγ target, via miR-331-5p) and protein phosphatase 1 regulatory subunit 3G (PPP1R3G, a Smad3 target). PPARγ knockdown/deletion in VSMCs activates TGFβ1 signaling. The PPARγ agonist pioglitazone reverses PAH and inhibits the TGFβ1-Stat3-FoxO1 axis in TGFβ1-overexpressing mice. We identified PPARγ as a missing link between BMP2 and TGFβ1 pathways in VSMCs. PPARγ activation can be beneficial in TGFβ1-associated diseases, such as PAH, parenchymal lung diseases, and Marfan's syndrome.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Nadine Hoffmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Jonas Geldner
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Paul Borchert
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover 30625, Germany
| | - Miklos M Mozes
- Department of Pathophysiology, Semmelweis University, Budapest 1089, Hungary
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
49
|
Hu C, Keen HL, Lu KT, Liu X, Wu J, Davis DR, Ibeawuchi SRC, Vogel S, Quelle FW, Sigmund CD. Retinol-binding protein 7 is an endothelium-specific PPAR γ cofactor mediating an antioxidant response through adiponectin. JCI Insight 2017; 2:e91738. [PMID: 28352663 PMCID: PMC5358481 DOI: 10.1172/jci.insight.91738] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Impaired PPARγ activity in endothelial cells causes oxidative stress and endothelial dysfunction which causes a predisposition to hypertension, but the identity of key PPARγ target genes that protect the endothelium remain unclear. Retinol-binding protein 7 (RBP7) is a PPARγ target gene that is essentially endothelium specific. Whereas RBP7-deficient mice exhibit normal endothelial function at baseline, they exhibit severe endothelial dysfunction in response to cardiovascular stressors, including high-fat diet and subpressor angiotensin II. Endothelial dysfunction was not due to differences in weight gain, impaired glucose homeostasis, or hepatosteatosis, but occurred through an oxidative stress-dependent mechanism which can be rescued by scavengers of superoxide. RNA sequencing revealed that RBP7 was required to mediate induction of a subset of PPARγ target genes by rosiglitazone in the endothelium including adiponectin. Adiponectin was selectively induced in the endothelium of control mice by high-fat diet and rosiglitazone, whereas RBP7 deficiency abolished this induction. Adiponectin inhibition caused endothelial dysfunction in control vessels, whereas adiponectin treatment of RBP7-deficient vessels improved endothelium-dependent relaxation and reduced oxidative stress. We conclude that RBP7 is required to mediate the protective effects of PPARγ in the endothelium through adiponectin, and RBP7 is an endothelium-specific PPARγ target and regulator of PPARγ activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Silke Vogel
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Curt D Sigmund
- Department of Pharmacology.,UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
50
|
Bai X, Mangum KD, Dee RA, Stouffer GA, Lee CR, Oni-Orisan A, Patterson C, Schisler JC, Viera AJ, Taylor JM, Mack CP. Blood pressure-associated polymorphism controls ARHGAP42 expression via serum response factor DNA binding. J Clin Invest 2017; 127:670-680. [PMID: 28112683 PMCID: PMC5272192 DOI: 10.1172/jci88899] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022] Open
Abstract
We recently demonstrated that selective expression of the Rho GTPase-activating protein ARHGAP42 in smooth muscle cells (SMCs) controls blood pressure by inhibiting RhoA-dependent contractility, providing a mechanism for the blood pressure-associated locus within the ARHGAP42 gene. The goals of the current study were to identify polymorphisms that affect ARHGAP42 expression and to better assess ARHGAP42's role in the development of hypertension. Using DNase I hypersensitivity methods and ENCODE data, we have identified a regulatory element encompassing the ARHGAP42 SNP rs604723 that exhibits strong SMC-selective, allele-specific activity. Importantly, CRISPR/Cas9-mediated deletion of this element in cultured human SMCs markedly reduced endogenous ARHGAP42 expression. DNA binding and transcription assays demonstrated that the minor T allele variation at rs604723 increased the activity of this fragment by promoting serum response transcription factor binding to a cryptic cis-element. ARHGAP42 expression was increased by cell stretch and sphingosine 1-phosphate in a RhoA-dependent manner, and deletion of ARHGAP42 enhanced the progression of hypertension in mice treated with DOCA-salt. Our analysis of a well-characterized cohort of untreated borderline hypertensive patients suggested that ARHGAP42 genotype has important implications in regard to hypertension risk. Taken together, our data add insight into the genetic mechanisms that control blood pressure and provide a potential target for individualized antihypertensive therapies.
Collapse
MESH Headings
- Animals
- Blood Pressure
- CRISPR-Cas Systems
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Gene Expression Regulation
- Humans
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Mice
- Mice, Transgenic
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Polymorphism, Single Nucleotide
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Sodium Chloride, Dietary/adverse effects
- Sodium Chloride, Dietary/pharmacology
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein/genetics
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
| | | | | | | | - Craig R. Lee
- McAllister Heart Institute, and
- Department of Pharmacy, University of North Carolina at Chapel Hill, Durham, North Carolina, USA
| | - Akinyemi Oni-Orisan
- Department of Pharmacy, University of North Carolina at Chapel Hill, Durham, North Carolina, USA
| | - Cam Patterson
- New York–Presbyterian Hospital/Weill Cornell Medical Center, New York, New York, USA
| | | | - Anthony J. Viera
- Department of Family Medicine, University of North Carolina at Chapel Hill, Durham, North Carolina, USA
| | | | | |
Collapse
|