1
|
Park M, Zhuang W, Jeong J, Kim HR, Jang Y, Seo MS, An JR, Park H, Han ET, Han JH, Chun W, Park WS. The SGLT2 inhibitor remogliflozin induces vasodilation in the femoral artery of rabbits via activation of a Kv channel, the SERCA pump, and the cGMP signaling pathway. Toxicol Appl Pharmacol 2025; 495:117228. [PMID: 39788209 DOI: 10.1016/j.taap.2025.117228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
This study explored the vasodilatory mechanisms of the sodium-glucose cotransporter-2 inhibitor remogliflozin using femoral arteries of rabbits. Remogliflozin dilated femoral arterial rings pre-contracted with phenylephrine in a concentration-dependent manner. Pretreatment with the Ca2+-sensitive K+ channel inhibitor (paxilline), the ATP-sensitive K+ channel inhibitor (glibenclamide), or the inwardly rectifying K+ channel inhibitor (Ba2+) did not alter the vasodilatory effect. However, vasodilation was significantly reduced by pretreatment with the voltage-dependent K+ (Kv) channel inhibitor (4-AP) and with the Kv1.5 subtype inhibitor (DPO-1) but not with Kv2.1 or Kv7 subtype inhibitor. Neither endothelium removal nor the inhibition of nitric oxide production altered the vasodilatory effect of remogliflozin. However, pretreatment with the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid effectively reduced the remogliflozin effect, as did pretreatment with cGMP/PKG-related but not cAMP/PKA-related signaling pathway inhibitors. These results indicate that remogliflozin-mediated dilation of the femoral artery occurs via the activation of Kv channels, mainly the Kv1.5 subtype, SERCA pump, and cGMP/PKG-related signaling pathways.
Collapse
Affiliation(s)
- Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Hye Ryung Kim
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - YeEun Jang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Mi Seon Seo
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, South Korea
| | - Jin Ryeol An
- Department of Physiology, KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 27478, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Eun-Taek Han
- Institute of Medical Sciences, Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Jin-Hee Han
- Institute of Medical Sciences, Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Wanjoo Chun
- Institute of Medical Sciences, Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea.
| |
Collapse
|
2
|
Kalyanaraman H, Casteel DE, Cabriales JA, Tat J, Zhuang S, Chan A, Dretchen KL, Boss GR, Pilz RB. The Antioxidant/Nitric Oxide-Quenching Agent Cobinamide Prevents Aortic Disease in a Mouse Model of Marfan Syndrome. JACC Basic Transl Sci 2024; 9:46-62. [PMID: 38362350 PMCID: PMC10864892 DOI: 10.1016/j.jacbts.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 02/17/2024]
Abstract
Major pathologic changes in the proximal aorta underlie the life-threatening aortic aneurysms and dissections in Marfan Syndrome; current treatments delay aneurysm development without addressing the primary pathology. Because excess oxidative stress and nitric oxide/protein kinase G signaling likely contribute to the aortopathy, we hypothesized that cobinamide, a strong antioxidant that can attenuate nitric oxide signaling, could be uniquely suited to prevent aortic disease. In a well-characterized mouse model of Marfan Syndrome, cobinamide dramatically reduced elastin breaks, prevented excess collagen deposition and smooth muscle cell apoptosis, and blocked DNA, lipid, and protein oxidation and excess nitric oxide/protein kinase G signaling in the ascending aorta. Consistent with preventing pathologic changes, cobinamide diminished aortic root dilation without affecting blood pressure. Cobinamide exhibited excellent safety and pharmacokinetic profiles indicating it could be a practical treatment. We conclude that cobinamide deserves further study as a disease-modifying treatment of Marfan Syndrome.
Collapse
Affiliation(s)
- Hema Kalyanaraman
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Darren E. Casteel
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Justin A. Cabriales
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - John Tat
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Adriano Chan
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | | | - Gerry R. Boss
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| | - Renate B. Pilz
- Department of Medicine, University of California-San Diego, La Jolla, California, USA
| |
Collapse
|
3
|
Shi F, Collins S. Regulation of mTOR Signaling: Emerging Role of Cyclic Nucleotide-Dependent Protein Kinases and Implications for Cardiometabolic Disease. Int J Mol Sci 2023; 24:11497. [PMID: 37511253 PMCID: PMC10380887 DOI: 10.3390/ijms241411497] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase is a central regulator of cell growth and metabolism. It is the catalytic subunit of two distinct large protein complexes, mTOR complex 1 (mTORC1) and mTORC2. mTOR activity is subjected to tight regulation in response to external nutrition and growth factor stimulation. As an important mechanism of signaling transduction, the 'second messenger' cyclic nucleotides including cAMP and cGMP and their associated cyclic nucleotide-dependent kinases, including protein kinase A (PKA) and protein kinase G (PKG), play essential roles in mediating the intracellular action of a variety of hormones and neurotransmitters. They have also emerged as important regulators of mTOR signaling in various physiological and disease conditions. However, the mechanism by which cAMP and cGMP regulate mTOR activity is not completely understood. In this review, we will summarize the earlier work establishing the ability of cAMP to dampen mTORC1 activation in response to insulin and growth factors and then discuss our recent findings demonstrating the regulation of mTOR signaling by the PKA- and PKG-dependent signaling pathways. This signaling framework represents a new non-canonical regulation of mTOR activity that is independent of AKT and could be a novel mechanism underpinning the action of a variety of G protein-coupled receptors that are linked to the mTOR signaling network. We will further review the implications of these signaling events in the context of cardiometabolic disease, such as obesity, non-alcoholic fatty liver disease, and cardiac remodeling. The metabolic and cardiac phenotypes of mouse models with targeted deletion of Raptor and Rictor, the two essential components for mTORC1 and mTORC2, will be summarized and discussed.
Collapse
Affiliation(s)
- Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Dhahri W, Dussault S, Raguema N, Desjarlais M, Rivard A. Stimulation of soluble guanylate cyclase activity with riociguat promotes angiogenesis and improves neovascularization after limb ischemia. Atherosclerosis 2023; 372:32-40. [PMID: 37023506 DOI: 10.1016/j.atherosclerosis.2023.03.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND AND AIMS The NO-cGMP pathway is essential for angiogenesis, vasculogenesis and post-natal neovascularization. The key enzyme responsible for the synthesis of cGMP following binding of NO is soluble guanylate cyclase (sGC). Riociguat is the first member of a novel class of compounds known as sGC stimulators. We tested the hypothesis that stimulation of sGC with riociguat might improve neovascularization in response to ischemia. METHODS In vitro, the angiogenic effect of riociguat was tested in human umbilical vein endothelial cells (HUVECs). In vivo, neovascularization was investigated in a mouse model of limb ischemia. C57Bl/6 mice were treated by gavage with 3 mg/kg/day of riociguat for a total of 28 days. After two weeks of treatment, hindlimb ischemia was surgically induced by femoral artery removal. RESULTS In a matrigel assay in vitro, riociguat dose-dependently stimulates tubule formation in HUVECs. Cell migration (scratch assay) is also increased in HUVECs treated with riociguat. At the molecular level, riociguat treatment leads to rapid activation of the p44/p42 MAP kinase pathway in HUVECs. Inhibition of protein kinase G (PKG) activity supresses both p44/p42 MAP kinase activation and angiogenesis in HUVECs treated with riociguat. In vivo, treatment with riociguat improves blood flow recovery after ischemia (Laser Doppler imaging), and increases capillary density in ischemic muscles (CD31 immunostaining). Clinically, this is associated with a significant decrease of ambulatory impairment and ischemic damages. Interestingly, mice treated with riociguat also show a 94% increase in the number of bone marrow-derived pro-angiogenic cells (PACs) compared to control mice. Moreover, riociguat treatment is associated with a significant improvement of PAC functions including migratory capacity, adhesion to an endothelial monolayer, and integration into endothelial tubular networks. CONCLUSIONS The sGC stimulator riociguat promotes angiogenesis and improves neovascularization after ischemia. The mechanism involves PKG-dependent activation of p44/p42 MAP kinase pathway, together with an improvement of PAC number and functions. sGC stimulation could constitute a novel therapeutic strategy to reduce tissue ischemia in patients with severe atherosclerotic diseases.
Collapse
Affiliation(s)
- Wahiba Dhahri
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Sylvie Dussault
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Nozha Raguema
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Michel Desjarlais
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada
| | - Alain Rivard
- Department of Medicine, Centre hospitalier de l'Université de Montréal (CHUM) Research Center, Montréal, Québec, Canada.
| |
Collapse
|
5
|
Targeting the crosstalk between canonical Wnt/β-catenin and inflammatory signaling cascades: A novel strategy for cancer prevention and therapy. Pharmacol Ther 2021; 227:107876. [PMID: 33930452 DOI: 10.1016/j.pharmthera.2021.107876] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Emerging scientific evidence indicates that inflammation is a critical component of tumor promotion and progression. Most cancers originate from sites of chronic irritation, infections and inflammation, underscoring that the tumor microenvironment is largely orchestrated by inflammatory cells and pro-inflammatory molecules. These inflammatory components are intimately involved in neoplastic processes which foster proliferation, survival, invasion, and migration, making inflammation the primary target for cancer prevention and treatment. The influence of inflammation and the immune system on the progression and development of cancer has recently gained immense interest. The Wnt/β-catenin signaling pathway, an evolutionarily conserved signaling strategy, has a critical role in regulating tissue development. It has been implicated as a major player in cancer development and progression with its regulatory role on inflammatory cascades. Many naturally-occurring and small synthetic molecules endowed with inherent anti-inflammatory properties inhibit this aberrant signaling pathway, making them a promising class of compounds in the fight against inflammatory cancers. This article analyzes available scientific evidence and suggests a crosslink between Wnt/β-catenin signaling and inflammatory pathways in inflammatory cancers, especially breast, gastrointestinal, endometrial, and ovarian cancer. We also highlight emerging experimental findings that numerous anti-inflammatory synthetic and natural compounds target the crosslink between Wnt/β-catenin pathway and inflammatory cascades to achieve cancer prevention and intervention. Current challenges, limitations, and future directions of research are also discussed.
Collapse
|
6
|
Gupta A, Behl T, Aleya L, Rahman MH, Yadav HN, Pal G, Kaur I, Arora S. Role of UPP pathway in amelioration of diabetes-associated complications. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:19601-19614. [PMID: 33660172 DOI: 10.1007/s11356-021-12781-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/29/2021] [Indexed: 06/12/2023]
Abstract
Type 2 diabetes (T2D) is one of the most widely spread metabolic disorder also called as "life style" disease. Due to the alarming number of patients, there is great need to therapies targeting functions which can help in maintaining the homeostasis of glucose levels and improving insulin sensitivity. Detailed analysis was done through various research and review papers which was searched using MEDLINE, BIOSIS, and EMBASE using various keywords. This search retrieved the most appropriate content on these molecules targeting UPP pathway. From this extensive review involving UPP pathway, it was concluded that the role of ubiquitin's is not only limited to neurodegenerative disorders but also plays a critical role in progression of diabetes including obesity, insulin resistance, and various neurogenerative disorders but it also targets proteasomal degradation including mediation of cellular signaling pathways. Thus, drugs targeting UPP not only may show effect against diabetes but also are therapeutically beneficial in the treatment of diabetes-associated complications which may be obtained. Thus, based on the available information and data on UPP functions, it can be concluded that regulation of UPP pathway via downstream regulators mainly E1, E2, and E3 may bring promising results. Drugs targeting these transcriptional factors may emerge as a novel therapy in the treatment of diabetes and diabetes-associated complications.
Collapse
Affiliation(s)
- Amit Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Lotfi Aleya
- Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, Besançon, France
| | - Md Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Seoul, South Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka, 1213, Bangladesh
| | | | - Giridhari Pal
- Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
7
|
Gorfe AA, Cho KJ. Approaches to inhibiting oncogenic K-Ras. Small GTPases 2021; 12:96-105. [PMID: 31438765 PMCID: PMC7849769 DOI: 10.1080/21541248.2019.1655883] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/29/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023] Open
Abstract
Activating somatic K-Ras mutations are associated with >15% all human tumors and up to 90% of specific tumor types such as pancreatic cancer. Successfully inhibiting abnormal K-Ras signaling would therefore be a game changer in cancer therapy. However, K-Ras has long been considered an undruggable target for various reasons. This view is now changing by the discovery of allosteric inhibitors that directly target K-Ras and inhibit its functions, and by the identification of new mechanisms to dislodge it from the plasma membrane and thereby abrogate its cellular activities. In this review, we will discuss recent progresses and challenges to inhibiting aberrant K-Ras functions by these two approaches. We will also provide a broad overview of other approaches such as inhibition of K-Ras effectors, and offer a brief perspective on the way forward.
Collapse
Affiliation(s)
- Alemayehu A. Gorfe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Programs of Biochemistry & Cell and Therapeutics & Pharmacology, MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Kwang-Jin Cho
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, USA
| |
Collapse
|
8
|
Mahneva O, Caplan SL, Ivko P, Dawson-Scully K, Milton SL. NO/cGMP/PKG activation protects Drosophila cells subjected to hypoxic stress. Comp Biochem Physiol C Toxicol Pharmacol 2019; 223:106-114. [PMID: 31150868 DOI: 10.1016/j.cbpc.2019.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 01/28/2023]
Abstract
The anoxia-tolerant fruit fly, Drosophila melanogaster, has routinely been used to examine cellular mechanisms responsible for anoxic and oxidative stress resistance. Nitric oxide (NO), an important cellular signaling molecule, and its downstream activation of cGMP-dependent protein kinase G (PKG) has been implicated as a protective mechanism against ischemic injury in diverse animal models from insects to mammals. In Drosophila, increased PKG signaling results in increased survival of animals exposed to anoxic stress. To determine if activation of the NO/cGMP/PKG pathway is protective at the cellular level, the present study employed a pharmacological protocol to mimic hypoxic injury in Drosophila S2 cells. The commonly used S2 cell line was derived from a primary culture of late stage (20-24 h old) Drosophila melanogaster embryos. Hypoxic stress was induced by exposure to either sodium azide (NaN3) or cobalt chloride (CoCl2). During chemical hypoxic stress, NO/cGMP/PKG activation protected against cell death and this mechanism involved modulation of downstream mitochondrial ATP-sensitive potassium ion channels (mitoKATP). The cellular protection afforded by NO/cGMP/PKG activation during ischemia-like stress may be an adaptive cytoprotective mechanism and modulation of this signaling cascade could serve as a potential therapeutic target for protection against hypoxia or ischemia-induced cellular injury.
Collapse
Affiliation(s)
- Olena Mahneva
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| | - Stacee Lee Caplan
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| | - Polina Ivko
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| | - Ken Dawson-Scully
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| | - Sarah L Milton
- Department of Biological Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431, USA.
| |
Collapse
|
9
|
Schwaerzer GK, Kalyanaraman H, Casteel DE, Dalton ND, Gu Y, Lee S, Zhuang S, Wahwah N, Schilling JM, Patel HH, Zhang Q, Makino A, Milewicz DM, Peterson KL, Boss GR, Pilz RB. Aortic pathology from protein kinase G activation is prevented by an antioxidant vitamin B 12 analog. Nat Commun 2019; 10:3533. [PMID: 31387997 PMCID: PMC6684604 DOI: 10.1038/s41467-019-11389-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 07/11/2019] [Indexed: 01/08/2023] Open
Abstract
People heterozygous for an activating mutation in protein kinase G1 (PRKG1, p.Arg177Gln) develop thoracic aortic aneurysms and dissections (TAAD) as young adults. Here we report that mice heterozygous for the mutation have a three-fold increase in basal protein kinase G (PKG) activity, and develop age-dependent aortic dilation. Prkg1R177Q/+ aortas show increased smooth muscle cell apoptosis, elastin fiber breaks, and oxidative stress compared to aortas from wild type littermates. Transverse aortic constriction (TAC)—to increase wall stress in the ascending aorta—induces severe aortic pathology and mortality from aortic rupture in young mutant mice. The free radical-neutralizing vitamin B12-analog cobinamide completely prevents age-related aortic wall degeneration, and the unrelated anti-oxidant N-acetylcysteine ameliorates TAC-induced pathology. Thus, increased basal PKG activity induces oxidative stress in the aorta, raising concern about the widespread clinical use of PKG-activating drugs. Cobinamide could be a treatment for aortic aneurysms where oxidative stress contributes to the disease, including Marfan syndrome. Individuals carrying a gain-of-function mutation in PKG1 develop thoracic aortic aneurysms and dissections. Here Schwaerzer et al. show that mice carrying the same mutation recapitulate the human disease, and find that treatment with anti-oxidants including cobinamide, a vitamin B12 analog, prevents disease progression.
Collapse
Affiliation(s)
- Gerburg K Schwaerzer
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hema Kalyanaraman
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Darren E Casteel
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nancy D Dalton
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Seunghoe Lee
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shunhui Zhuang
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nisreen Wahwah
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jan M Schilling
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hemal H Patel
- Department of Anesthesiology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Qian Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ayako Makino
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Dianna M Milewicz
- Division of Medical Genetics and Cardiology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Kirk L Peterson
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Gerry R Boss
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Renate B Pilz
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Lehners M, Dobrowinski H, Feil S, Feil R. cGMP Signaling and Vascular Smooth Muscle Cell Plasticity. J Cardiovasc Dev Dis 2018; 5:jcdd5020020. [PMID: 29671769 PMCID: PMC6023364 DOI: 10.3390/jcdd5020020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
Cyclic GMP regulates multiple cell types and functions of the cardiovascular system. This review summarizes the effects of cGMP on the growth and survival of vascular smooth muscle cells (VSMCs), which display remarkable phenotypic plasticity during the development of vascular diseases, such as atherosclerosis. Recent studies have shown that VSMCs contribute to the development of atherosclerotic plaques by clonal expansion and transdifferentiation to macrophage-like cells. VSMCs express a variety of cGMP generators and effectors, including NO-sensitive guanylyl cyclase (NO-GC) and cGMP-dependent protein kinase type I (cGKI), respectively. According to the traditional view, cGMP inhibits VSMC proliferation, but this concept has been challenged by recent findings supporting a stimulatory effect of the NO-cGMP-cGKI axis on VSMC growth. Here, we summarize the relevant studies with a focus on VSMC growth regulation by the NO-cGMP-cGKI pathway in cultured VSMCs and mouse models of atherosclerosis, restenosis, and angiogenesis. We discuss potential reasons for inconsistent results, such as the use of genetic versus pharmacological approaches and primary versus subcultured cells. We also explore how modern methods for cGMP imaging and cell tracking could help to improve our understanding of cGMP’s role in vascular plasticity. We present a revised model proposing that cGMP promotes phenotypic switching of contractile VSMCs to VSMC-derived plaque cells in atherosclerotic lesions. Regulation of vascular remodeling by cGMP is not only an interesting new therapeutic strategy, but could also result in side effects of clinically used cGMP-elevating drugs.
Collapse
Affiliation(s)
- Moritz Lehners
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Hyazinth Dobrowinski
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Susanne Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
11
|
Wang SB, Venkatraman V, Crowgey EL, Liu T, Fu Z, Holewinski R, Ranek M, Kass DA, O'Rourke B, Van Eyk JE. Protein S-Nitrosylation Controls Glycogen Synthase Kinase 3β Function Independent of Its Phosphorylation State. Circ Res 2018; 122:1517-1531. [PMID: 29563102 DOI: 10.1161/circresaha.118.312789] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/13/2018] [Accepted: 03/19/2018] [Indexed: 01/11/2023]
Abstract
RATIONALE GSK-3β (glycogen synthase kinase 3β) is a multifunctional and constitutively active kinase known to regulate a myriad of cellular processes. The primary mechanism to regulate its function is through phosphorylation-dependent inhibition at serine-9 residue. Emerging evidence indicates that there may be alternative mechanisms that control GSK-3β for certain functions. OBJECTIVES Here, we sought to understand the role of protein S-nitrosylation (SNO) on the function of GSK-3β. SNO-dependent modulation of the localization of GSK-3β and its ability to phosphorylate downstream targets was investigated in vitro, and the network of proteins differentially impacted by phospho- or SNO-dependent GSK-3β regulation and in vivo SNO modification of key signaling kinases during the development of heart failure was also studied. METHODS AND RESULTS We found that GSK-3β undergoes site-specific SNO both in vitro, in HEK293 cells, H9C2 myoblasts, and primary neonatal rat ventricular myocytes, as well as in vivo, in hearts from an animal model of heart failure and sudden cardiac death. S-nitrosylation of GSK-3β significantly inhibits its kinase activity independent of the canonical phospho-inhibition pathway. S-nitrosylation of GSK-3β promotes its nuclear translocation and access to novel downstream phosphosubstrates which are enriched for a novel amino acid consensus sequence motif. Quantitative phosphoproteomics pathway analysis reveals that nuclear GSK-3β plays a central role in cell cycle control, RNA splicing, and DNA damage response. CONCLUSIONS The results indicate that SNO has a differential effect on the location and activity of GSK-3β in the cytoplasm versus the nucleus. SNO modification of GSK-3β occurs in vivo and could contribute to the pathobiology of heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Sheng-Bing Wang
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Vidya Venkatraman
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Erin L Crowgey
- Department of Nemours Biomedical Research, Nemours Alfred I. duPont Hospital for Children, Wilmington, DE (E.L.C.)
| | - Ting Liu
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | | | - Ronald Holewinski
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.).,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| | - Mark Ranek
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - David A Kass
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Brian O'Rourke
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.)
| | - Jennifer E Van Eyk
- From the Department of Medicine (S.-B.W., V.V., T.L., R.H., M.R., D.A.K., B.O'R., J.E.V.E.) .,Johns Hopkins University, Baltimore, MD; Department of Medicine, Advanced Clinical Biosystems Research Institute, The Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (V.V., R.H., J.E.V.E.)
| |
Collapse
|
12
|
Gao Q, Tang J, Li N, Zhou X, Zhu X, Li W, Liu B, Feng X, Tao J, Han B, Zhang H, Sun M, Xu Z. New conception for the development of hypertension in preeclampsia. Oncotarget 2018; 7:78387-78395. [PMID: 27861155 PMCID: PMC5346647 DOI: 10.18632/oncotarget.13410] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/14/2016] [Indexed: 12/27/2022] Open
Abstract
Placental vascular dysfunction was suggested to be critical for placental ischemia-initiated hypertension in preeclampsia, although the contributions of endothelium involved are unclear. The present study found, unlike non-placental vessels, acetylcholine showed no vasodilatation effect on placental vessels, indicating that endothelial-derived nitric oxide (NO) was extremely weak in placental vessels. Placental vascular responses to exogenous NO from sodium nitroprusside (SNP) were significantly different from non-placental vessels. These results were further confirmed in sheep, and rat vessels. In preeclamptic placental vessels, acetylcholine also showed no vasodilatation effects, while vascular responses to SNP were suppressed, associated with impaired cGMP/sGC pathway in vascular smooth muscle cells (VSMCs). The current theory on placental ischemia-initiated hypertension in preeclampsia focused on changes in placental vascular functions, including endothelial dysfunction. This study found the placental endothelium contributed very poorly to vasodilatation, and altered vascular functions in preeclampsia mainly occurred in VSMCs instead of endothelial cells. The findings contribute importantly to understanding the special feature of placental vascular functions and its pathophysiological changes in the development of hypertension in preeclampsia.
Collapse
Affiliation(s)
- Qinqin Gao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaolin Zhu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weisheng Li
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Bailin Liu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueqin Feng
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianying Tao
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Obstetrics and Gynecology, First and Second Affiliated Hospital of Soochow University, Municipal Hospital, Suzhou, China
| | - Bing Han
- Department of Obstetrics and Gynecology, First and Second Affiliated Hospital of Soochow University, Municipal Hospital, Suzhou, China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, First and Second Affiliated Hospital of Soochow University, Municipal Hospital, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Affiliated Hospital of Soochow University, Suzhou, China.,Center for Perinatal Biology, Loma Linda University, California, USA
| |
Collapse
|
13
|
Jiao C, Zhu L, Gu Z. GSK-3 mediates NO-cGMP-induced isoflavone production in soybean sprouts. Food Res Int 2017; 101:203-208. [PMID: 28941685 DOI: 10.1016/j.foodres.2017.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/19/2017] [Accepted: 09/03/2017] [Indexed: 10/18/2022]
Abstract
The role of glycogen synthase kinase-3 (GSK-3) in the nitric oxide-guanosine 3',5'-cyclic monophosphate (NO-cGMP)-induced isoflavone production in soybean sprouts was examined. Inhibitors and donors of NO, cGMP, and GSK-3 inhibitor were added to UV-B irradiated sprouts. Results showed that NO, with cGMP, induced the expression of GSK-3 under UV-B radiation. Protein kinase G (PKG) was shown to be involved in NO-cGMP-induced GSK-3 activation. GSK-3 elevated activity and expression levels of chalcone synthase (CHS) and isoflavone synthase (IFS), and increased isoflavone accumulation.
Collapse
Affiliation(s)
- Caifeng Jiao
- College of Life Science, Anqing Normal University, Anqing, Anhui 246133, People's Republic of China.
| | - Liangliang Zhu
- College of Life Science, Anqing Normal University, Anqing, Anhui 246133, People's Republic of China
| | - Zhenxin Gu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| |
Collapse
|
14
|
Gao Q, Tang J, Li N, Liu B, Zhang M, Sun M, Xu Z. What is precise pathophysiology in development of hypertension in pregnancy? Precision medicine requires precise physiology and pathophysiology. Drug Discov Today 2017; 23:286-299. [PMID: 29101000 DOI: 10.1016/j.drudis.2017.10.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/25/2017] [Accepted: 10/23/2017] [Indexed: 01/12/2023]
Abstract
It is widely accepted that placental ischemia is central in the evolution of hypertension in pregnancy. Many studies and reviews have targeted placental ischemia to explain mechanisms for initiating pregnancy hypertension. The placenta is rich in blood vessels, which are the basis for developing placental ischemia. However, is the physiology of placental vessels the same as that of nonplacental vessels? What is the pathophysiology of placental vessels in development of pregnancy hypertension? This review aims to provide a comprehensive summary of special features of placental vascular regulations and the pathophysiological changes linked to preeclamptic conditions. Interestingly, some popular theories or accepted concepts could be based on our limited knowledge and evidence regarding placental vascular physiology, pharmacology and pathophysiology. New views raised could offer interesting ideas for future investigation of mechanisms as well as targets for pregnancy hypertension.
Collapse
Affiliation(s)
- Qinqin Gao
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Bailin Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Mengshu Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China.
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China; Center for Perinatal Biology, Loma Linda University, CA, USA.
| |
Collapse
|
15
|
AMPK and Endothelial Nitric Oxide Synthase Signaling Regulates K-Ras Plasma Membrane Interactions via Cyclic GMP-Dependent Protein Kinase 2. Mol Cell Biol 2016; 36:3086-3099. [PMID: 27697864 DOI: 10.1128/mcb.00365-16] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
K-Ras must localize to the plasma membrane and be arrayed in nanoclusters for biological activity. We show here that K-Ras is a substrate for cyclic GMP-dependent protein kinases (PKGs). In intact cells, activated PKG2 selectively colocalizes with K-Ras on the plasma membrane and phosphorylates K-Ras at Ser181 in the C-terminal polybasic domain. K-Ras phosphorylation by PKG2 is triggered by activation of AMP-activated protein kinase (AMPK) and requires endothelial nitric oxide synthase and soluble guanylyl cyclase. Phosphorylated K-Ras reorganizes into distinct nanoclusters that retune the signal output. Phosphorylation acutely enhances K-Ras plasma membrane affinity, but phosphorylated K-Ras is progressively lost from the plasma membrane via endocytic recycling. Concordantly, chronic pharmacological activation of AMPK → PKG2 signaling with mitochondrial inhibitors, nitric oxide, or sildenafil inhibits proliferation of K-Ras-positive non-small cell lung cancer cells. The study shows that K-Ras is a target of a metabolic stress-signaling pathway that can be leveraged to inhibit oncogenic K-Ras function.
Collapse
|
16
|
Roberts JL, Tavallai M, Nourbakhsh A, Fidanza A, Cruz-Luna T, Smith E, Siembida P, Plamondon P, Cycon KA, Doern CD, Booth L, Dent P. GRP78/Dna K Is a Target for Nexavar/Stivarga/Votrient in the Treatment of Human Malignancies, Viral Infections and Bacterial Diseases. J Cell Physiol 2015; 230:2552-78. [PMID: 25858032 PMCID: PMC4843173 DOI: 10.1002/jcp.25014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 01/10/2023]
Abstract
Prior tumor cell studies have shown that the drugs sorafenib (Nexavar) and regorafenib (Stivarga) reduce expression of the chaperone GRP78. Sorafenib/regorafenib and the multi‐kinase inhibitor pazopanib (Votrient) interacted with sildenafil (Viagra) to further rapidly reduce GRP78 levels in eukaryotes and as single agents to reduce Dna K levels in prokaryotes. Similar data were obtained in tumor cells in vitro and in drug‐treated mice for: HSP70, mitochondrial HSP70, HSP60, HSP56, HSP40, HSP10, and cyclophilin A. Prolonged ‘rafenib/sildenafil treatment killed tumor cells and also rapidly decreased the expression of: the drug efflux pumps ABCB1 and ABCG2; and NPC1 and NTCP, receptors for Ebola/Hepatitis A and B viruses, respectively. Pre‐treatment with the ‘Rafenib/sildenafil combination reduced expression of the Coxsackie and Adenovirus receptor in parallel with it also reducing the ability of a serotype 5 Adenovirus or Coxsackie virus B4 to infect and to reproduce. Sorafenib/pazopanib and sildenafil was much more potent than sorafenib/pazopanib as single agents at preventing Adenovirus, Mumps, Chikungunya, Dengue, Rabies, West Nile, Yellow Fever, and Enterovirus 71 infection and reproduction. ‘Rafenib drugs/pazopanib as single agents killed laboratory generated antibiotic resistant E. coli which was associated with reduced Dna K and Rec A expression. Marginally toxic doses of ‘Rafenib drugs/pazopanib restored antibiotic sensitivity in pan‐antibiotic resistant bacteria including multiple strains of blakpcKlebsiella pneumoniae. Thus, Dna K is an antibiotic target for sorafenib, and inhibition of GRP78/Dna K has therapeutic utility for cancer and for bacterial and viral infections. J. Cell. Physiol. 230: 2552–2578, 2015. © 2015 The Authors. Journal of Cellular Physiology published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Aida Nourbakhsh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | - Christopher D Doern
- Department of Pathology, Virginia Commonwealth University, Richmond, Virginia
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
17
|
Tavallai M, Hamed HA, Roberts JL, Cruickshanks N, Chuckalovcak J, Poklepovic A, Booth L, Dent P. Nexavar/Stivarga and viagra interact to kill tumor cells. J Cell Physiol 2015; 230:2281-98. [PMID: 25704960 PMCID: PMC4835179 DOI: 10.1002/jcp.24961] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 12/29/2022]
Abstract
We determined whether the multi‐kinase inhibitor sorafenib or its derivative regorafenib interacted with phosphodiesterase 5 (PDE5) inhibitors such as Viagra (sildenafil) to kill tumor cells. PDE5 and PDGFRα/β were over‐expressed in liver tumors compared to normal liver tissue. In multiple cell types in vitro sorafenib/regorafenib and PDE5 inhibitors interacted in a greater than additive fashion to cause tumor cell death, regardless of whether cells were grown in 10 or 100% human serum. Knock down of PDE5 or of PDGFRα/β recapitulated the effects of the individual drugs. The drug combination increased ROS/RNS levels that were causal in cell killing. Inhibition of CD95/FADD/caspase 8 signaling suppressed drug combination toxicity. Knock down of ULK‐1, Beclin1, or ATG5 suppressed drug combination lethality. The drug combination inactivated ERK, AKT, p70 S6K, and mTOR and activated JNK. The drug combination also reduced mTOR protein expression. Activation of ERK or AKT was modestly protective whereas re‐expression of an activated mTOR protein or inhibition of JNK signaling almost abolished drug combination toxicity. Sildenafil and sorafenib/regorafenib interacted in vivo to suppress xenograft tumor growth using liver and colon cancer cells. From multiplex assays on tumor tissue and plasma, we discovered that increased FGF levels and ERBB1 and AKT phosphorylation were biomarkers that were directly associated with lower levels of cell killing by ‘rafenib + sildenafil. Our data are now being translated into the clinic for further determination as to whether this drug combination is a useful anti‐tumor therapy for solid tumor patients. J. Cell. Physiol. 230: 2281–2298, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mehrad Tavallai
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Hossein A Hamed
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Nichola Cruickshanks
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
18
|
Kim YJ, Ibrahim LA, Wang SZ, Yuan W, Evgrafov OV, Knowles JA, Wang K, Tao HW, Zhang LI. EphA7 regulates spiral ganglion innervation of cochlear hair cells. Dev Neurobiol 2015; 76:452-69. [PMID: 26178595 DOI: 10.1002/dneu.22326] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/10/2015] [Accepted: 07/14/2015] [Indexed: 01/11/2023]
Abstract
During the development of periphery auditory circuitry, spiral ganglion neurons (SGNs) form a spatially precise pattern of innervation of cochlear hair cells (HCs), which is an essential structural foundation for central auditory processing. However, molecular mechanisms underlying the developmental formation of this precise innervation pattern remain not well understood. Here, we specifically examined the involvement of Eph family members in cochlear development. By performing RNA-sequencing for different types of cochlear cell, in situ hybridization, and immunohistochemistry, we found that EphA7 was strongly expressed in a large subset of SGNs. In EphA7 deletion mice, there was a reduction in the number of inner radial bundles originating from SGNs and projecting to HCs as well as in the number of ribbon synapses on inner hair cells (IHCs), as compared with wild-type or heterozygous mutant mice, attributable to fewer type I afferent fibers. The overall activity of the auditory nerve in EphA7 deletion mice was also reduced, although there was no significant change in the hearing intensity threshold. In vitro analysis further suggested that the reduced innervation of HCs by SGNs could be attributed to a role of EphA7 in regulating outgrowth of SGN neurites as knocking down EphA7 in SGNs resulted in diminished SGN fibers. In addition, suppressing the activity of ERK1/2, a potential downstream target of EphA7 signaling, either with specific inhibitors in cultured explants or by knocking out Prkg1, also resulted in reduced SGN fibers. Together, our results suggest that EphA7 plays an important role in the developmental formation of cochlear innervation pattern through controlling SGN fiber ontogeny. Such regulation may contribute to the salience level of auditory signals presented to the central auditory system.
Collapse
Affiliation(s)
- Young J Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Neuroscience Graduate Program, University Of Southern California, Los Angeles, California
| | - Leena A Ibrahim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Neuroscience Graduate Program, University Of Southern California, Los Angeles, California
| | - Sheng-Zhi Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033
| | - Wei Yuan
- Department of Otolaryngology of Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Oleg V Evgrafov
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - James A Knowles
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Kai Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Cell And Neurobiology, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Physiology and Biophysics, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| |
Collapse
|
19
|
Booth L, Roberts JL, Tavallai M, Nourbakhsh A, Chuckalovcak J, Carter J, Poklepovic A, Dent P. OSU-03012 and Viagra Treatment Inhibits the Activity of Multiple Chaperone Proteins and Disrupts the Blood-Brain Barrier: Implications for Anti-Cancer Therapies. J Cell Physiol 2015; 230:1982-98. [PMID: 25736380 PMCID: PMC4835175 DOI: 10.1002/jcp.24977] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 02/26/2015] [Indexed: 12/14/2022]
Abstract
We examined the interaction between OSU‐03012 (also called AR‐12) with phosphodiesterase 5 (PDE5) inhibitors to determine the role of the chaperone glucose‐regulated protein (GRP78)/BiP/HSPA5 in the cellular response. Sildenafil (Viagra) interacted in a greater than additive fashion with OSU‐03012 to kill stem‐like GBM cells. Treatment of cells with OSU‐03012/sildenafil: abolished the expression of multiple oncogenic growth factor receptors and plasma membrane drug efflux pumps and caused a rapid degradation of GRP78 and other HSP70 and HSP90 family chaperone proteins. Decreased expression of plasma membrane receptors and drug efflux pumps was dependent upon enhanced PERK‐eIF2α‐ATF4‐CHOP signaling and was blocked by GRP78 over‐expression. In vivo OSU‐03012/sildenafil was more efficacious than treatment with celecoxib and sildenafil at killing tumor cells without damaging normal tissues and in parallel reduced expression of ABCB1 and ABCG2 in the normal brain. The combination of OSU‐03012/sildenafil synergized with low concentrations of sorafenib to kill tumor cells, and with lapatinib to kill ERBB1 over‐expressing tumor cells. In multiplex assays on plasma and human tumor tissue from an OSU‐03012/sildenafil treated mouse, we noted a profound reduction in uPA signaling and identified FGF and JAK1/2 as response biomarkers for potentially suppressing the killing response. Inhibition of FGFR signaling and to a lesser extent JAK1/2 signaling profoundly enhanced OSU‐03012/sildenafil lethality. J. Cell. Physiol. 230: 1982–1998, 2015. © 2015 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Booth L, Roberts JL, Cash DR, Tavallai S, Jean S, Fidanza A, Cruz-Luna T, Siembiba P, Cycon KA, Cornelissen CN, Dent P. GRP78/BiP/HSPA5/Dna K is a universal therapeutic target for human disease. J Cell Physiol 2015; 230:1661-76. [PMID: 25546329 PMCID: PMC4402027 DOI: 10.1002/jcp.24919] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/18/2014] [Indexed: 01/11/2023]
Abstract
The chaperone GRP78/Dna K is conserved throughout evolution down to prokaryotes. The GRP78 inhibitor OSU-03012 (AR-12) interacted with sildenafil (Viagra) or tadalafil (Cialis) to rapidly reduce GRP78 levels in eukaryotes and as a single agent reduce Dna K levels in prokaryotes. Similar data with the drug combination were obtained for: HSP70, HSP90, GRP94, GRP58, HSP27, HSP40 and HSP60. OSU-03012/sildenafil treatment killed brain cancer stem cells and decreased the expression of: NPC1 and TIM1; LAMP1; and NTCP1, receptors for Ebola/Marburg/Hepatitis A, Lassa fever, and Hepatitis B viruses, respectively. Pre-treatment with OSU-03012/sildenafil reduced expression of the coxsakie and adenovirus receptor in parallel with it also reducing the ability of a serotype 5 adenovirus or coxsakie virus B4 to infect and to reproduce. Similar data were obtained using Chikungunya, Mumps, Measles, Rubella, RSV, CMV, and Influenza viruses. OSU-03012 as a single agent at clinically relevant concentrations killed laboratory generated antibiotic resistant E. coli and clinical isolate multi-drug resistant N. gonorrhoeae and MRSE which was in bacteria associated with reduced Dna K and Rec A expression. The PDE5 inhibitors sildenafil or tadalafil enhanced OSU-03012 killing in N. gonorrhoeae and MRSE and low marginally toxic doses of OSU-03012 could restore bacterial sensitivity in N. gonorrhoeae to multiple antibiotics. Thus, Dna K and bacterial phosphodiesterases are novel antibiotic targets, and inhibition of GRP78 is of therapeutic utility for cancer and also for bacterial and viral infections. J. Cell. Physiol. 230: 1661–1676, 2015. © 2014 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Booth L, Roberts JL, Cruickshanks N, Tavallai S, Webb T, Samuel P, Conley A, Binion B, Young HF, Poklepovic A, Spiegel S, Dent P. PDE5 inhibitors enhance celecoxib killing in multiple tumor types. J Cell Physiol 2015; 230:1115-27. [PMID: 25303541 DOI: 10.1002/jcp.24843] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/02/2014] [Indexed: 12/20/2022]
Abstract
The present studies determined whether clinically relevant phosphodiesterase 5 (PDE5) inhibitors interacted with a clinically relevant NSAID, celecoxib, to kill tumor cells. Celecoxib and PDE5 inhibitors interacted in a greater than additive fashion to kill multiple tumor cell types. Celecoxib and sildenafil killed ex vivo primary human glioma cells as well as their associated activated microglia. Knock down of PDE5 recapitulated the effects of PDE5 inhibitor treatment; the nitric oxide synthase inhibitor L-NAME suppressed drug combination toxicity. The effects of celecoxib were COX2 independent. Over-expression of c-FLIP-s or knock down of CD95/FADD significantly reduced killing by the drug combination. CD95 activation was dependent on nitric oxide and ceramide signaling. CD95 signaling activated the JNK pathway and inhibition of JNK suppressed cell killing. The drug combination inactivated mTOR and increased the levels of autophagy and knock down of Beclin1 or ATG5 strongly suppressed killing by the drug combination. The drug combination caused an ER stress response; knock down of IRE1α/XBP1 enhanced killing whereas knock down of eIF2α/ATF4/CHOP suppressed killing. Sildenafil and celecoxib treatment suppressed the growth of mammary tumors in vivo. Collectively our data demonstrate that clinically achievable concentrations of celecoxib and sildenafil have the potential to be a new therapeutic approach for cancer.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Park S, Kim JK, Oh CJ, Choi SH, Jeon JH, Lee IK. Scoparone interferes with STAT3-induced proliferation of vascular smooth muscle cells. Exp Mol Med 2015; 47:e145. [PMID: 25744297 PMCID: PMC4351406 DOI: 10.1038/emm.2014.113] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/29/2014] [Accepted: 11/06/2014] [Indexed: 01/15/2023] Open
Abstract
Scoparone, which is a major constituent of Artemisia capillaries, has been identified as an anticoagulant, hypolipidemic, vasorelaxant, anti-oxidant and anti-inflammatory drug, and it is used for the traditional treatment of neonatal jaundice. Therefore, we hypothesized that scoparone could suppress the proliferation of VSMCs by interfering with STAT3 signaling. We found that the proliferation of these cells was significantly attenuated by scoparone in a dose-dependent manner. Scoparone markedly reduced the serum-stimulated accumulation of cells in the S phase and concomitantly increased the proportion of cells in the G0/G1 phase, which was consistent with the reduced expression of cyclin D1, phosphorylated Rb and survivin in the VSMCs. Cell adhesion markers, such as MCP-1 and ICAM-1, were significantly reduced by scoparone. Interestingly, this compound attenuated the increase in cyclin D promoter activity by inhibiting the activities of both the WT and active forms of STAT3. Similarly, the expression of a cell proliferation marker induced by PDGF was decreased by scoparone with no change in the phosphorylation of JAK2 or Src. On the basis of the immunofluorescence staining results, STAT3 proteins phosphorylated by PDGF were predominantly localized to the nucleus and were markedly reduced in the scoparone-treated cells. In summary, scoparone blocks the accumulation of STAT3 transported from the cytosol to the nucleus, leading to the suppression of VSMC proliferation through G1 phase arrest and the inhibition of Rb phosphorylation. This activity occurs independent of the form of STAT3 and upstream of kinases, such as Jak and Src, which are correlated with abnormal vascular remodeling due to the presence of an excess of growth factors following vascular injury. These data provide convincing evidence that scoparone may be a new preventative agent for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Sungmi Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jeong-Kook Kim
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Joo Oh
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Hee Choi
- Department of Biomedical Science, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Han Jeon
- Departments of Internal Medicine, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- 1] Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea [2] BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea [3] Department of Biomedical Science, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea [4] Departments of Internal Medicine, Kyungpook National University School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
23
|
Chen J, Roberts JD. cGMP-dependent protein kinase I gamma encodes a nuclear localization signal that regulates nuclear compartmentation and function. Cell Signal 2014; 26:2633-44. [PMID: 25172423 PMCID: PMC4254301 DOI: 10.1016/j.cellsig.2014.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/15/2014] [Indexed: 10/24/2022]
Abstract
cGMP-dependent protein kinase I (PKGI) plays an important role in regulating how cGMP specifies vascular smooth muscle cell (SMC) phenotype. Although studies indicate that PKGI nuclear localization controls how cGMP regulates gene expression in SMC, information about the mechanisms that regulate PKGI nuclear compartmentation and its role in directly regulating cell phenotype is limited. Here we characterize a nuclear localization signal sequence (NLS) in PKGIγ, a proteolytically cleaved PKGI kinase fragment that translocates to the nucleus of SMC. Immuno-localization studies using cells expressing native and NLS-mutant PKGIγ, and treated with a small molecule nuclear transport inhibitor, indicated that PKGIγ encodes a constitutively active NLS that requires importin α and β for regulation of its compartmentation. Moreover, studies utilizing a genetically encoded nuclear phospho-CREB biosensor probe and fluorescence lifetime imaging microscopy demonstrated that this NLS controls PKGIγ nuclear function. In addition, although cytosolic PKGIγ-activity was observed to stimulate MAPK/ERK-mediated nuclear CREB signaling in SMC, NLS-mediated PKGIγ nuclear activity alone was determined to increase the expression of differentiation marker proteins in these cells. These results indicate that NLS-mediated nuclear PKGIγ localization plays an important role in how PKGI regulates vascular SMC phenotype.
Collapse
Affiliation(s)
- Jingsi Chen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA
| | - Jesse D Roberts
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA; Departments of Anesthesia, Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
24
|
Pandey KN. Guanylyl cyclase/natriuretic peptide receptor-A signaling antagonizes phosphoinositide hydrolysis, Ca(2+) release, and activation of protein kinase C. Front Mol Neurosci 2014; 7:75. [PMID: 25202235 PMCID: PMC4141235 DOI: 10.3389/fnmol.2014.00075] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 08/05/2014] [Indexed: 11/25/2022] Open
Abstract
Thus far, three related natriuretic peptides (NPs) and three distinct sub-types of cognate NP receptors have been identified and characterized based on the specific ligand binding affinities, guanylyl cyclase activity, and generation of intracellular cGMP. Atrial and brain natriuretic peptides (ANP and BNP) specifically bind and activate guanylyl cyclase/natriuretic peptide receptor-A (GC-A/NPRA), and C-type natriuretic peptide (CNP) shows specificity to activate guanylyl cyclase/natriuretic peptide receptor-B (GC-B/NPRB). All three NPs bind to natriuretic peptide receptor-C (NPRC), which is also known as clearance or silent receptor. The NPRA is considered the principal biologically active receptor of NP family; however, the molecular signaling mechanisms of NP receptors are not well understood. The activation of NPRA and NPRB produces the intracellular second messenger cGMP, which serves as the major signaling molecule of all three NPs. The activation of NPRB in response to CNP also produces the intracellular cGMP; however, at lower magnitude than that of NPRA, which is activated by ANP and BNP. In addition to enhanced accumulation of intracellular cGMP in response to all three NPs, the levels of cAMP, Ca2+ and inositol triphosphate (IP3) have also been reported to be altered in different cells and tissue types. Interestingly, ANP has been found to lower the concentrations of cAMP, Ca2+, and IP3; however, NPRC has been proposed to increase the levels of these metabolic signaling molecules. The mechanistic studies of decreased and/or increased levels of cAMP, Ca2+, and IP3 in response to NPs and their receptors have not yet been clearly established. This review focuses on the signaling mechanisms of ANP/NPRA and their biological effects involving an increased level of intracellular accumulation of cGMP and a decreased level of cAMP, Ca2+, and IP3 in different cells and tissue systems.
Collapse
Affiliation(s)
- Kailash N Pandey
- Department of Physiology, School of Medicine, Tulane University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
25
|
Booth L, Roberts JL, Cruickshanks N, Grant S, Poklepovic A, Dent P. Regulation of OSU-03012 toxicity by ER stress proteins and ER stress-inducing drugs. Mol Cancer Ther 2014; 13:2384-98. [PMID: 25103559 DOI: 10.1158/1535-7163.mct-14-0172] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The present studies examined the toxic interaction between the non-coxib celecoxib derivative OSU-03012 and phosphodiesterase 5 (PDE5) inhibitors, and also determined the roles of endoplasmic reticulum stress response regulators in cell survival. PDE5 inhibitors interacted in a greater than additive fashion with OSU-03012 to kill parental glioma and stem-like glioma cells. Knockdown of the endoplasmic reticulum stress response proteins IRE1 or XBP1 enhanced the lethality of OSU-03012, and of [OSU-03012 + PDE5 inhibitor] treatment. Pan-caspase and caspase-9 inhibition did not alter OSU-03012 lethality but did abolish enhanced killing in the absence of IRE1 or XBP1. Expression of the mitochondrial protective protein BCL-XL or the caspase-8 inhibitor c-FLIP-s, or knockdown of death receptor CD95 or the death receptor caspase-8 linker protein FADD, suppressed killing by [OSU-03012 + PDE5 inhibitor] treatment. CD95 activation was blocked by the nitric oxide synthase inhibitor L-NAME. Knockdown of the autophagy regulatory proteins Beclin1 or ATG5 protected the cells from OSU-03012 and from [OSU-03012 + PDE5 inhibitor] toxicity. Knockdown of IRE1 enhanced OSU-03012/[OSU-03012 + PDE5 inhibitor]-induced JNK activation, and inhibition of JNK suppressed the elevated killing caused by IRE1 knockdown. Knockdown of CD95 blunted JNK activation. Collectively, our data demonstrate that PDE5 inhibitors recruit death receptor signaling to enhance OSU-03012 toxicity in glioblastoma multiforme (GBM) cells.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jane L Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Nichola Cruickshanks
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Steven Grant
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Andrew Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
26
|
Zhang J, Chen J, Xu C, Yang J, Guo Q, Hu Q, Jiang H. Resveratrol inhibits phenotypic switching of neointimal vascular smooth muscle cells after balloon injury through blockade of Notch pathway. J Cardiovasc Pharmacol 2014; 63:233-239. [PMID: 24603118 DOI: 10.1097/fjc.0000000000000040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells (VSMCs) plays an initial role in neointimal hyperplasia, the main cause of many occlusive vascular diseases. The aim of this study was to measure the effects of resveratrol (RSV) on the phenotypic transformation of VSMCs and to investigate its mechanism of action. METHODS Cultured VSMCs isolated from rat thoracic aorta were prepared with serum starvation for 72 hours followed by RSV treatment (50-200 μmol/L) and 10% serum stimulation. Male Sprague-Dawley rats, subjected to carotid arteries injury from a balloon catheter, were exposed to intraperitoneal injection of RSV (1 mg/kg) or saline and were killed after 7 or 28 days. RESULTS Compared with cells in the serum-induced group, VSMCs in the RSV or N-[N-(3, 5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) treatment group exhibited significant decreases of proliferation and migration. The total and cytoplasmic Notch-1 levels were declined by RSV, accompanied by a significant increase in smooth muscle α-actin and smooth muscle myosin heavy chain protein. The expression of Notch-1, Jagged-1, Hey-1, and Hey-2 mRNA in balloon-injured arteries at 7 days was decreased by RSV treatment. Arteries from RSV-treated rats showed less neointimal hyperplasia, lower collagen content, and a lower rate of cells positive for proliferating cell nuclear antigen 28 days after injury, compared with saline controls. CONCLUSIONS The results indicate that RSV can attenuate phenotypic switching of VSMCs after arterial injury through inhibition of the Notch pathway.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Aorta, Thoracic/cytology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/pathology
- Carotid Arteries/drug effects
- Carotid Arteries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cells, Cultured
- Disease Models, Animal
- Hyperplasia/prevention & control
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Neointima/prevention & control
- Rats
- Rats, Sprague-Dawley
- Receptor, Notch1/genetics
- Resveratrol
- Stilbenes/pharmacology
- Time Factors
Collapse
Affiliation(s)
- Jing Zhang
- *Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; †Departments of Cardiology; and ‡Ophthalmology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Zhang X, Kainz V, Zhao J, Strassman AM, Levy D. Vascular extracellular signal-regulated kinase mediates migraine-related sensitization of meningeal nociceptors. Ann Neurol 2013; 73:741-50. [PMID: 23447360 DOI: 10.1002/ana.23873] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 01/04/2013] [Accepted: 02/15/2013] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To examine changes in the response properties of meningeal nociceptors that might lead to migraine pain and examine endogenous processes that could play a role in mediating them using a clinically relevant model of migraine triggering, namely infusion of the nitric oxide (NO) donor nitroglycerin (NTG). METHODS Single-unit recordings made in the trigeminal ganglion of rats were used to test changes in the activity and mechanosensitivity of meningeal nociceptors in response to administration of the migraine trigger NTG or another NO donor S-nitroso-N-acetyl-DL-penicillamine (SNAP) at doses relevant to the human model of migraine headache. Immunohistochemistry and pharmacological manipulations were used to investigate the possible role of meningeal vascular signaling in mediating the responses of meningeal nociceptors to NO. RESULTS Infusion of NTG promoted a delayed and robust increase in the mechanosensitivity of meningeal nociceptors, with a time course resembling the development of the delayed migraine headache. A similar sensitization was elicited by dural application of NTG and SNAP. NTG-evoked delayed meningeal nociceptor sensitization was associated with a robust extracellular signal-regulated kinase (ERK) phosphorylation in meningeal arteries. Pharmacological blockade of meningeal ERK phosphorylation inhibited the development of NTG-evoked delayed meningeal nociceptor sensitization. INTERPRETATION The development of delayed mechanical sensitization evoked by the migraine trigger NTG is potentially of great importance as the first finding of a neurophysiological correlate of migraine headache in meningeal nociceptors. The arterial ERK phosphorylation and its involvement in mediating the NTG-evoked delayed sensitization points to an important, yet unappreciated, role of the meningeal vasculature in the genesis of migraine pain.
Collapse
Affiliation(s)
- XiChun Zhang
- Departments of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | |
Collapse
|
28
|
Chen CN, Watson G, Zhao L. Cyclic guanosine monophosphate signalling pathway in pulmonary arterial hypertension. Vascul Pharmacol 2012; 58:211-8. [PMID: 22982057 DOI: 10.1016/j.vph.2012.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 08/28/2012] [Accepted: 09/04/2012] [Indexed: 12/19/2022]
Abstract
During the last decade, it emerged that cyclic guanosine monophosphate (cGMP) is a novel drug target for the treatment of pulmonary arterial hypertension (PAH). cGMP regulates many cellular functions, ranging from contractility to growth, of relevance to the disease. Generated from guanylyl cyclases in response to natriuretic peptides or nitric oxide (NO), cGMP transduces its effects through a number of cGMP effectors, including cGMP-regulated phosphodiesterases and protein kinases. Furthermore, the cGMP concentration is modulated by cGMP-degrading phosphodiesterases. Data to date demonstrate that increasing intracellular cGMP through stimulation of GCs, inhibition of PDEs, or both is a valid therapeutic strategy in drug development for PAH. New advances in understanding of cGMP are unravelled, as well as the pathobiology of PAH. cGMP remains an attractive future PAH drug target. This review makes a more detailed examination of cGMP signalling with particular reference to PAH.
Collapse
Affiliation(s)
- Chien-nien Chen
- Experimental Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | | | | |
Collapse
|
29
|
Ochoa CD, Alexeyev M, Pastukh V, Balczon R, Stevens T. Pseudomonas aeruginosa exotoxin Y is a promiscuous cyclase that increases endothelial tau phosphorylation and permeability. J Biol Chem 2012; 287:25407-18. [PMID: 22637478 DOI: 10.1074/jbc.m111.301440] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Exotoxin Y (ExoY) is a type III secretion system effector found in ~ 90% of the Pseudomonas aeruginosa isolates. Although it is known that ExoY causes inter-endothelial gaps and vascular leak, the mechanisms by which this occurs are poorly understood. Using both a bacteria-delivered and a codon-optimized conditionally expressed ExoY, we report that this toxin is a dual soluble adenylyl and guanylyl cyclase that results in intracellular cAMP and cGMP accumulation. The enzymatic activity of ExoY caused phosphorylation of endothelial Tau serine 214, accumulation of insoluble Tau, inter-endothelial cell gap formation, and increased macromolecular permeability. To discern whether the cAMP or cGMP signal was responsible for Tau phosphorylation and barrier disruption, pulmonary microvascular endothelial cells were engineered for the conditional expression of either wild-type guanylyl cyclase, which synthesizes cGMP, or a mutated guanylyl cyclase, which synthesizes cAMP. Sodium nitroprusside stimulation of the cGMP-generating cyclase resulted in transient Tau serine 214 phosphorylation and gap formation, whereas stimulation of the cAMP-generating cyclase induced a robust increase in Tau serine 214 phosphorylation, gap formation, and macromolecular permeability. These results indicate that the cAMP signal is the dominant stimulus for Tau phosphorylation. Hence, ExoY is a promiscuous cyclase and edema factor that uses cAMP and, to some extent, cGMP to induce the hyperphosphorylation and insolubility of endothelial Tau. Because hyperphosphorylated and insoluble Tau are hallmarks in neurodegenerative tauopathies such as Alzheimer disease, acute Pseudomonas infections cause a pathophysiological sequela in endothelium previously recognized only in chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Cristhiaan D Ochoa
- Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | |
Collapse
|
30
|
Babykutty S, Suboj P, Srinivas P, Nair AS, Chandramohan K, Gopala S. Insidious role of nitric oxide in migration/invasion of colon cancer cells by upregulating MMP-2/9 via activation of cGMP-PKG-ERK signaling pathways. Clin Exp Metastasis 2012; 29:471-92. [PMID: 22419013 DOI: 10.1007/s10585-012-9464-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/26/2012] [Indexed: 12/16/2022]
Abstract
Nitric oxide (NO), an uncharged free radical is implicated in various physiological and pathological processes. The present study is an investigation on the effect of NO on proliferation, apoptosis and migration of colon cancer cells. Colon adenocarcinoma cells, WiDr, were used for the in vitro experiments. Tissues from colon adenocarcinoma, adjacent normal and inflammatory tissue and lymph node with metastasis were evaluated for iNOS, MMP-2/9 and Fra-1/Fra-2. NO increases the proliferation of cancer cells and simultaneously prevents apoptosis. Expression of MMP-2/9, RhoB and Rac-1 was enhanced by NO in a time dependent manner. Further, NO increased phosphorylation of ERK1/2 and induced nuclear translocation of Fra-1 and Fra-2. Electrophoretic mobility shift analysis and use of deletion mutant promoter constructs identified role of AP-1 in NO-mediated regulation of MMP-2/9. iNOS, MMP-2/9, Fra-1 and Fra-2 in normal and colon adenocarcinoma tissues were analyzed and it was found that increased expression of these proteins in cancer when compared to normal provides support to our in vitro findings. The study showed that the NO-cGMP-PKG promotes MMP-2/9 expression by activating ERK-1/2 and AP-1. This study reveals the insidious role of NO in imparting tumor aggressiveness.
Collapse
Affiliation(s)
- Suboj Babykutty
- Department of Biochemistry, Sree Chitra Tirunal Institute for Medical Sciences and Technology, 695011, Thiruvananthapuram, Kerala, India
| | | | | | | | | | | |
Collapse
|
31
|
cGMP-Prkg1 signaling and Pde5 inhibition shelter cochlear hair cells and hearing function. Nat Med 2012; 18:252-9. [PMID: 22270721 DOI: 10.1038/nm.2634] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 12/07/2011] [Indexed: 02/07/2023]
Abstract
Noise-induced hearing loss (NIHL) is a global health hazard with considerable pathophysiological and social consequences that has no effective treatment. In the heart, lung and other organs, cyclic guanosine monophosphate (cGMP) facilitates protective processes in response to traumatic events. We therefore analyzed NIHL in mice with a genetic deletion of the gene encoding cGMP-dependent protein kinase type I (Prkg1) and found a greater vulnerability to and markedly less recovery from NIHL in these mice as compared to mice without the deletion. Prkg1 was expressed in the sensory cells and neurons of the inner ear of wild-type mice, and its expression partly overlapped with the expression profile of cGMP-hydrolyzing phosphodiesterase 5 (Pde5). Treatment of rats and wild-type mice with the Pde5 inhibitor vardenafil almost completely prevented NIHL and caused a Prkg1-dependent upregulation of poly (ADP-ribose) in hair cells and the spiral ganglion, suggesting an endogenous protective cGMP-Prkg1 signaling pathway that culminates in the activation of poly (ADP-ribose) polymerase. These data suggest vardenafil or related drugs as possible candidates for the treatment of NIHL.
Collapse
|
32
|
Kotlo KU, Hesabi B, Danziger RS. Implication of microRNAs in atrial natriuretic peptide and nitric oxide signaling in vascular smooth muscle cells. Am J Physiol Cell Physiol 2011; 301:C929-37. [PMID: 21734186 DOI: 10.1152/ajpcell.00088.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRs) are endogenous small RNA molecules that suppress gene expression by binding to complementary sequences in the 3' untranslated regions of their target genes. miRs have been implicated in many diseases, including heart failure, ischemic heart disease, hypertension, cardiac hypertrophy, and cancers. Nitric oxide (NO) and atrial natriuretic peptide (ANP) are potent vasorelaxants whose actions are mediated through receptor guanylyl cyclases and cGMP-dependent protein kinase. The present study examines miRs in signaling by ANP and NO in vascular smooth muscle cells. miR microarray analysis was performed on human vascular smooth muscle cells (HVSMC) treated with ANP (10 nM, 4 h) and S-nitroso-N-acetylpenicillamine (SNAP) (100 μM, 4 h), a NO donor. Twenty-two shared miRs were upregulated, and 21 shared miRs were downregulated, by both ANP and SNAP (P < 0.05). Expression levels of four miRs (miRs-21, -26b, -98, and -1826), which had the greatest change in expression, as determined by microarray analysis, were confirmed by quantitative RT-PCR. Rp-8-Br-PET-cGMPS, a cGMP-dependent protein kinase-specific inhibitor, blocked the regulation of these miRs by ANP and SNAP. 8-bromo-cGMP mimicked the effect of ANP and SNAP on their expression. miR-21 was shown to inhibit HVSMC contraction in collagen gel lattice contraction assays. We also identified by computational algorithms and confirmed by Western blot analysis new intracellular targets of miR-21, i.e., cofilin-2 and myosin phosphatase and Rho interacting protein. Transfection with pre-miR-21 contracted cells and ANP and SNAP blocked miR-21-induced HVSMC contraction. Transfection with anti-miR-21 inhibitor reduced contractility of HVSMC (P < 0.05). The present results implicate miRs in NO and ANP signaling in general and miR-21 in particular in cGMP signaling and vascular smooth muscle cell relaxation.
Collapse
Affiliation(s)
- Kumar U Kotlo
- Department of Medicine, University of Illinois at Chicago, 60612, USA.
| | | | | |
Collapse
|
33
|
cGMP-dependent protein kinases as potential targets for colon cancer prevention and treatment. Future Med Chem 2011; 2:65-80. [PMID: 21426046 DOI: 10.4155/fmc.09.142] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, several antitumor signaling pathways mediated by the cGMP-dependent protein kinases have been identified in colon cancer cells. This review aims to present the mounting evidence in favor of cGMP/protein kinase G (PKG) signaling as a therapeutic strategy in colon cancer. The homeostatic and tumor suppressive effects of cGMP in the intestine are uncontested, but the signaling details are not understood. PKG is the central cGMP effector, and can block proliferation and tumor angiogenesis by inhibiting β-catenin/TCF and SOX9 signaling. Therapeutic activation of cGMP/PKG offers a promising avenue for the prevention and treatment of colon cancer, but additional preclinical studies are needed to fully understand the potential of this system.
Collapse
|
34
|
Doronzo G, Viretto M, Russo I, Mattiello L, Di Martino L, Cavalot F, Anfossi G, Trovati M. Nitric oxide activates PI3-K and MAPK signalling pathways in human and rat vascular smooth muscle cells: Influence of insulin resistance and oxidative stress. Atherosclerosis 2011; 216:44-53. [DOI: 10.1016/j.atherosclerosis.2011.01.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 01/11/2011] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
|
35
|
Korkmaz B, Buharalioglu K, Sahan-Firat S, Cuez T, Tuncay Demiryurek A, Tunctan B. Activation of MEK1/ERK1/2/iNOS/sGC/PKG pathway associated with peroxynitrite formation contributes to hypotension and vascular hyporeactivity in endotoxemic rats. Nitric Oxide 2011; 24:160-72. [DOI: 10.1016/j.niox.2011.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2010] [Revised: 12/11/2010] [Accepted: 02/20/2011] [Indexed: 01/09/2023]
|
36
|
Acute Lung Injury: The Injured Lung Endothelium, Therapeutic Strategies for Barrier Protection, and Vascular Biomarkers. TEXTBOOK OF PULMONARY VASCULAR DISEASE 2010. [PMCID: PMC7120335 DOI: 10.1007/978-0-387-87429-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
37
|
Protein kinase G activity prevents pathological-level nitric oxide-induced apoptosis and promotes DNA synthesis/cell proliferation in vascular smooth muscle cells. Cardiovasc Pathol 2010; 19:e221-31. [DOI: 10.1016/j.carpath.2009.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2009] [Revised: 08/26/2009] [Accepted: 11/02/2009] [Indexed: 11/19/2022] Open
|
38
|
Lamon BD, Upmacis RK, Deeb RS, Koyuncu H, Hajjar DP. Inducible nitric oxide synthase gene deletion exaggerates MAPK-mediated cyclooxygenase-2 induction by inflammatory stimuli. Am J Physiol Heart Circ Physiol 2010; 299:H613-23. [PMID: 20543082 DOI: 10.1152/ajpheart.00144.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cyclooxygenase (COX)-2 and inducible nitric oxide (NO) synthase (iNOS) are responsive to a wide array of inflammatory stimuli, have been localized to vascular smooth muscle cells (SMCs), and are intimately linked to the progression of vascular disease, including atherosclerotic lesion formation. We and others have shown that the production and subsequent impact of COX products appear to be correlative with the status of NO synthesis. This study examined the impact of inflammation-driven NO production on COX-2 expression in SMCs. Concurrent stimulation of quiescent rat aortic SMCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma increased COX-2, iNOS, and nitrite production. Pharmacological inhibition of NO synthase (N(G)-monomethyl-l-arginine) concentration- and time-dependently magnified LPS + IFN-gamma-mediated COX-2 mRNA and protein induction in a cGMP-independent manner. COX-2 induction was associated with activation of the ERK, p38, and JNK mitogen-activated protein kinase (MAPK) pathways. Interestingly, NO synthase inhibition enhanced ERK, p38, and to a lesser extent JNK phosphorylation but suppressed MAPK phosphatase (MKP)-1 induction in response to LPS + IFN-gamma. Similarly, the exposure of SMCs from iNOS(-/-) mice to LPS + IFN-gamma produced an enhancement of COX-2 induction, p38, and JNK phosphorylation and an attenuated upregulation of MKP-1 versus their wild-type counterparts. Taken together, our data indicate that NO, in part derived from iNOS, negatively regulates the immediate early induction of COX-2 in response to inflammatory stimuli.
Collapse
Affiliation(s)
- Brian D Lamon
- Department of Pathology and Laboratory Medicine, Center of Vascular Biology, Weill Cornell Medical College of CornellUniversity, New York, New York 10065, USA.
| | | | | | | | | |
Collapse
|
39
|
Gratzke C, Angulo J, Chitaley K, Dai YT, Kim NN, Paick JS, Simonsen U, Uckert S, Wespes E, Andersson KE, Lue TF, Stief CG. Anatomy, physiology, and pathophysiology of erectile dysfunction. J Sex Med 2010; 7:445-75. [PMID: 20092448 DOI: 10.1111/j.1743-6109.2009.01624.x] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Significant scientific advances during the past 3 decades have deepened our understanding of the physiology and pathophysiology of penile erection. A critical evaluation of the current state of knowledge is essential to provide perspective for future research and development of new therapies. AIM To develop an evidence-based, state-of-the-art consensus report on the anatomy, physiology, and pathophysiology of erectile dysfunction (ED). METHODS Consensus process over a period of 16 months, representing the opinions of 12 experts from seven countries. MAIN OUTCOME MEASURE Expert opinion was based on the grading of scientific and evidence-based medical literature, internal committee discussion, public presentation, and debate. RESULTS ED occurs from multifaceted, complex mechanisms that can involve disruptions in neural, vascular, and hormonal signaling. Research on central neural regulation of penile erection is progressing rapidly with the identification of key neurotransmitters and the association of neural structures with both spinal and supraspinal pathways that regulate sexual function. In parallel to advances in cardiovascular physiology, the most extensive efforts in the physiology of penile erection have focused on elucidating mechanisms that regulate the functions of the endothelium and vascular smooth muscle of the corpus cavernosum. Major health concerns such as atherosclerosis, hyperlipidemia, hypertension, diabetes, and metabolic syndrome (MetS) have become well integrated into the investigation of ED. CONCLUSIONS Despite the efficacy of current therapies, they remain insufficient to address growing patient populations, such as those with diabetes and MetS. In addition, increasing awareness of the adverse side effects of commonly prescribed medications on sexual function provides a rationale for developing new treatment strategies that minimize the likelihood of causing sexual dysfunction. Many basic questions with regard to erectile function remain unanswered and further laboratory and clinical studies are necessary.
Collapse
Affiliation(s)
- Christian Gratzke
- Department of Urology, Ludwig-Maximilians-Universität, München, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Metukuri MR, Namas R, Gladstone C, Clermont T, Jefferson B, Barclay D, Hermus L, Billiar TR, Zamora R, Vodovotz Y. Activation of latent transforming growth factor-beta1 by nitric oxide in macrophages: role of soluble guanylate cyclase and MAP kinases. Wound Repair Regen 2009; 17:578-88. [PMID: 19614923 DOI: 10.1111/j.1524-475x.2009.00509.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The inducible nitric oxide (NO) synthase and the cytokine transforming growth factor-beta1 (TGF-beta1), both central modulators of wound healing, interact reciprocally: TGF-beta1 generally suppresses iNOS expression, while NO can induce and activate latent TGF-beta1. We have shown that chemical NO activates recombinant human latent TGF-beta1 by S-nitrosation of the latency-associated peptide (LAP), a cleaved portion of pro-TGF-beta1 that maintains TGF-beta1 in a biologically-inactive state. We hypothesized that cell-associated TGF-beta1 could be activated by NO via known NO-inducible signaling pathways (soluble guanylate cyclase [sGC] and mitogen-activated protein [MAP] kinases). Treatment of mouse RAW 264.7 macrophage-like cells with the NO donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP) led to a dose- and time-dependent increase in cell-associated active and latent TGF-beta1, as assessed by quantitative immunocytochemistry for active TGF-beta1 vs. LAP and partially validated by western blot analysis. Treatment with the sGC inhibitor 1,H-[1,2,4]oxadiazole[4,3-a]quinoxalon-1-one (ODQ) reduced both active and latent TGF-beta1 dose-dependently. SNAP, in the presence or absence of ODQ or the MAP kinase inhibitors, did not affect steady-state TGF-beta1 mRNA levels. Treatment with inhibitors specific for JNK1/2, ERK1/2, and p38 MAP kinases suppressed SNAP-induced active and latent TGF-beta1. Treatment with the cell-permeable cGMP analog 8-Br-cGMP increased both active and latent TGF-beta1. However, TGF-beta1 activation induced by 8-Br-cGMP was not blocked by MAP kinase inhibitors. Our findings suggest that NO activates latent TGF-beta1 via activation of sGC and generation of cGMP and separately via MAP kinase activation, and may shed insight into the mechanisms by which both cGMP production and MAP kinase activation enhance wound healing.
Collapse
|
41
|
Das A, Xi L, Kukreja RC. Protein kinase G-dependent cardioprotective mechanism of phosphodiesterase-5 inhibition involves phosphorylation of ERK and GSK3beta. J Biol Chem 2008; 283:29572-85. [PMID: 18723505 DOI: 10.1074/jbc.m801547200] [Citation(s) in RCA: 161] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sildenafil, a potent inhibitor of phosphodiesterase-5 (PDE-5) induces powerful protection against myocardial ischemia-reperfusion injury. PDE-5 inhibition increases cGMP levels that activate cGMP-dependent protein kinase (PKG). However, the cause and effect relationship of PKG in sildenafil-induced cardioprotection and the downstream targets of PKG remain unclear. Adult ventricular myocytes were treated with sildenafil and subjected to simulated ischemia and reoxygenation. Sildenafil treatment significantly decreased cardiomyocyte necrosis and apoptosis. The PKG inhibitors, KT5823, guanosine 3',5'-cyclic monophosphorothioate, 8-(4-chloro-phenylthio) (R(p)-8-pCPT-cGMPs), or DT-2 blocked the anti-necrotic and anti-apoptotic effect of sildenafil. Selective knockdown of PKG in cardiomyocytes with adenoviral vector containing short hairpin RNA of PKG also abolished sildenafil-induced protection. Furthermore, intra-coronary infusion of sildenafil in Langendorff-isolated mouse hearts prior to ischemia-reperfusion significantly reduced myocardial infarct size after 20 min ischemia and 30 min reperfusion, which was abrogated by KT5823. Sildenafil significantly increased PKG activity in intact hearts and cardiomyocytes. Sildenafil also enhanced the Bcl-2/Bax ratio, phosphorylation of Akt, ERK1/2, and glycogen synthase kinase 3beta. All these changes (except Akt phosphorylation) were significantly blocked by KT5823 and short hairpin RNA of PKG. These studies provide the first evidence for an essential role of PKG in sildenafil-induced cardioprotection. Moreover, our results demonstrate that sildenafil activates a PKG-dependent novel signaling cascade that involves activation of ERK and inhibition of glycogen synthase kinase 3beta leading to cytoprotection.
Collapse
Affiliation(s)
- Anindita Das
- Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | |
Collapse
|
42
|
Weinmeister P, Lukowski R, Linder S, Traidl-Hoffmann C, Hengst L, Hofmann F, Feil R. Cyclic guanosine monophosphate-dependent protein kinase I promotes adhesion of primary vascular smooth muscle cells. Mol Biol Cell 2008; 19:4434-41. [PMID: 18685080 DOI: 10.1091/mbc.e08-04-0370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The cyclic guanosine monophosphate (cGMP)/cGMP-dependent protein kinase type I (cGKI) pathway regulates many cellular functions. The current study shows that 8-Br-cGMP stimulates the number of attached primary but not that of subcultured murine vascular smooth muscle cells (VSMCs). These effects of 8-Br-cGMP require the presence of cGKI. In agreement with previous studies, cGKI inhibited the number of cells in repeatedly passaged murine VSMCs. Activation of the cGMP/cGKI pathway in freshly isolated primary VSMCs slightly decreased apoptosis and strongly increased cell adhesion. The stimulation of cell adhesion by cGKI involves an inhibition of the RhoA/Rho kinase pathway and increased exposure of beta(1) and beta(3) integrins on the cell surface. Together, these results identify a novel proadhesive function of cGMP/cGKI signaling in primary VSMCs and suggest that the opposing effects of this pathway on VSMC number depend on the phenotypic context of the cells.
Collapse
Affiliation(s)
- Pascal Weinmeister
- Institut für Pharmakologie und Toxikologie, Technischen Universiät München, D-80802 München, Germany.
| | | | | | | | | | | | | |
Collapse
|
43
|
Chien WL, Liang KC, Fu WM. Enhancement of active shuttle avoidance response by the NO-cGMP-PKG activator YC-1. Eur J Pharmacol 2008; 590:233-40. [PMID: 18590724 DOI: 10.1016/j.ejphar.2008.06.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2008] [Revised: 05/23/2008] [Accepted: 06/08/2008] [Indexed: 10/21/2022]
Abstract
Although much has been learned about the role of the amygdala in Pavlovian fear conditioning, relatively little is known about the signaling pathway involved in the acquisition of an active avoidance reaction. The aim of this study is to investigate the potentiating effects of the NO-guanylate cyclase activator YC-1 on learning and memory of shuttle avoidance test in rats. YC-1 enhanced the induction of long-term potentiation (LTP) in amygdala through NO-cGMP-PKG-ERK pathway and the increase of BDNF expression. The Western blot and PCR methods were used to examine the signaling pathways involved in fear memory. It was found that YC-1 increased the avoidance responses during learning period and the memory retention lasted longer than one week. The enhancement of learning behavior by YC-1 was antagonized by intracerebroventricular injection of NOS inhibitor l-NAME, PKG inhibitor Rp-8-Br-PET-cGMPS and MEK inhibitor PD98059, indicating that NO-cGMP-PKG and ERK pathways are involved in the learning potentiating action of YC-1. In addition, YC-1 increased the activation of ERK and Akt 30 min after Day-1 training in amygdala. YC-1 also potentiated the expression of BDNF and CREB in response to fear memory test. Taken together, these findings suggest that NO-cGMP-PKG-ERK signaling pathway is involved in the action of YC-1 in enhancing the fear memory.
Collapse
Affiliation(s)
- Wei-Lin Chien
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | |
Collapse
|
44
|
Browning DD. Protein kinase G as a therapeutic target for the treatment of metastatic colorectal cancer. Expert Opin Ther Targets 2008; 12:367-76. [PMID: 18269345 DOI: 10.1517/14728222.12.3.367] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Colorectal cancer is a leading cause of cancer-related death in the world and there is an urgent need for new strategies to combat this disease. Findings from several independent laboratories have converged on cGMP signaling as an exciting new therapeutic target, but the mechanisms remain controversial. A key intracellular effector of cGMP is protein kinase G (PKG). This article reviews the scientific literature concerning PKG effects on tumor development and progression, and discusses possible strategies for its exploitation in future cancer therapies. Studies from several independent laboratories have described novel anti-tumor effects of PKG in colon cancer cells that include inhibition of tumor growth and angiogenesis. While more preclinical research is warranted to better understand signaling mechanisms, these properties support the notion that PKG is a novel cancer target.
Collapse
Affiliation(s)
- Darren D Browning
- Medical College of Georgia, Department of Biochemistry and Molecular Biology, 1120 15th Street, CB2605, Augusta, GA 30912-2100, USA.
| |
Collapse
|
45
|
Scott A, Khan KM, Duronio V, Hart DA. Mechanotransduction in human bone: in vitro cellular physiology that underpins bone changes with exercise. Sports Med 2008; 38:139-60. [PMID: 18201116 PMCID: PMC3951486 DOI: 10.2165/00007256-200838020-00004] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bone has a remarkable ability to adjust its mass and architecture in response to a wide range of loads, from low-level gravitational forces to high-level impacts. A variety of types and magnitudes of mechanical stimuli have been shown to influence human bone cell metabolism in vitro, including fluid shear, tensile and compressive strain, altered gravity and vibration. Therefore, the current article aims to synthesize in vitro data regarding the cellular mechanisms underlying the response of human bone cells to mechanical loading. Current data demonstrate commonalities in response to different types of mechanical stimuli on the one hand, along with differential activation of intracellular signalling on the other. A major unanswered question is, how do bone cells sense and distinguish between different types of load? The studies included in the present article suggest that the type and magnitude of loading may be discriminated by overlapping mechanosensory mechanisms including (i) ion channels; (ii) integrins; (iii) G-proteins; and (iv) the cytoskeleton. The downstream signalling pathways identified to date appear to overlap with known growth factor and hormone signals, providing a mechanism of interaction between systemic influences and the local mechanical environment. Finally, the data suggest that exercise should emphasize the amount of load rather than the number of repetitions.
Collapse
Affiliation(s)
- Alexander Scott
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | |
Collapse
|
46
|
Marfella R, D' Amico M, Di Filippo C, Siniscalchi M, Sasso FC, Ferraraccio F, Rossi F, Paolisso G. The possible role of the ubiquitin proteasome system in the development of atherosclerosis in diabetes. Cardiovasc Diabetol 2007; 6:35. [PMID: 17971205 PMCID: PMC2169213 DOI: 10.1186/1475-2840-6-35] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 10/30/2007] [Indexed: 12/17/2022] Open
Abstract
We have reviewed the impact of the ubiquitin proteasome system (UPS) on atherosclerosis progression of diabetic patients. A puzzle of many pieces of evidence suggests that UPS, in addition to its role in the removal of damaged proteins, is involved in a number of biological processes including inflammation, proliferation and apoptosis, all of which constitute important characteristics of atherosclerosis. From what can be gathered from the very few studies on the UPS in diabetic cardiovascular diseases published so far, the system seems to be functionally active to a different extent in the initiation, progression, and complication stage of atherosclerosis in the diabetic people. Further evidence for this theory, however, has to be given, for instance by specifically targeted antagonism of the UPS. Nonetheless, this hypothesis may help us understand why diverse therapeutic interventions, which have in common the ability to reduce ubiquitin-proteasome activity, can impede or delay the onset of diabetes and cardiovascular diseases (CVD). People with type 2 diabetes are disproportionately affected by CVD, compared with those without diabetes [1]. The prevalence, incidence, and mortality from all forms of CVD (myocardial infarction, cerebro-vascular disease and congestive heart failure) are strikingly increased in persons with diabetes compared with those withoutdiabetes [2]. Furthermore, diabetic patients have not benefited by the advances in the management of obesity, dyslipidemia, and hypertension that have resulted in a decrease in mortality for coronary heart disease (CHD) patients without diabetes [3]. Nevertheless, these risk factors do not fully explain the excess risk for CHD associated with diabetes [4,5]. Thus, the determinants of progression of atherosclerosis in persons with diabetes must be elucidated. Beyond the major risk factors, several studies have demonstrated that such factors, strictly related to diabetes, as insulin-resistance, post-prandial hyperglycemia and chronic hyperglycemia play a role in the atherosclerotic process and may require intervention [6,7]. Moreover, it is important to recognize that these risk factors frequently "cluster" inindividual patients and possibly interact with each other, favouring the atherosclerosis progression toward plaque instability. Thus, a fundamental question is, "which is the common soil hypothesis that may unifying the burden of all these factors on atherosclerosis of diabetic patients? Because evidences suggest that insulin-resistance, diabetes and CHD share in common a deregulation of ubiquitin-proteasome system (UPS), the major pathway for nonlysosomal intracellular protein degradation in eucaryotic cells [8,9], in this review ubiquitin-proteasome deregulation is proposed as the common persistent pathogenic factor mediating the initial stage of the atherosclerosis as well as the progression to complicated plaque in diabetic patients.
Collapse
Affiliation(s)
- Raffaele Marfella
- Department of Geriatrics and Metabolic Diseases, Second University of Naples, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Gomes ER, Almeida RD, Carvalho AP, Duarte CB. Nitric Oxide Modulates Tumor Cell Death Induced by Photodynamic Therapy Through a cGMP-dependent Mechanism¶. Photochem Photobiol 2007. [DOI: 10.1562/0031-8655(2002)0760423nomtcd2.0.co2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
48
|
Sosa V, Carbó R, Guarner V. Participation of glucose transporters on atrial natriuretic peptide-induced glucose uptake by adult and neonatal cardiomyocytes under oxygenation and hypoxia. Eur J Pharmacol 2007; 568:83-8. [PMID: 17537429 DOI: 10.1016/j.ejphar.2007.04.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2007] [Revised: 04/18/2007] [Accepted: 04/20/2007] [Indexed: 11/22/2022]
Abstract
Natriuretic peptides, beside their endocrine actions, have paracrine functions which include regulating glucose uptake and metabolism. Atrial natriuretic peptide (ANP) actions are mediated by cGMP which is implicated in the metabolic adaptation of glucose metabolism to oxygen deprivation in the heart. Although, it has been reported that ANP increases glucose uptake, cGMP decreases it. The aim of the present paper was to evaluate the role of the glucose transporters 1 and 4 (GLUTS), in glucose uptake produced by ANP in fatty acid-dependent adult cardiomyocytes and glucose-dependent neonatal cardiomyocytes under oxygenation and hypoxia, which reverts adult metabolism to glucose-dependent. We also explored if the calcium-calmodulin complex participates in ANP-induced increase in glucose uptake. Neonatal cells had a higher glucose uptake than adult cells and GLUT 1 participated in basal uptake in both cell types. Hypoxia increased glucose uptake in adult cardiomyocytes but not in neonatal cells and this increase in glucose uptake was mediated by GLUT4. ANP increased glucose uptake in both adult and neonatal myocytes, under oxygenation and hypoxia, and GLUT4 favored this increase. Neonatal cells were less sensitive to ANP. Trifluoperazine, a calcium-calmodulin blocker, inhibited the ANP-induced increase in glucose uptake. This suggests that ANP promotes GLUT 4 calcium-mediated recruitment to the cell membrane. In conclusion, glucose uptake regulation is one of the paracrine metabolic effects of ANP in adult and neonatal cardiomyocytes under oxygenation and hypoxia. This effect of this peptide could explain the beneficial effects found in the internal medicine and surgical fields.
Collapse
Affiliation(s)
- Verónica Sosa
- Physiology Department National Institute of Cardiology Ignacio Chávez, Mexico D.F
| | | | | |
Collapse
|
49
|
Etgen AM, González-Flores O, Todd BJ. The role of insulin-like growth factor-I and growth factor-associated signal transduction pathways in estradiol and progesterone facilitation of female reproductive behaviors. Front Neuroendocrinol 2006; 27:363-75. [PMID: 16904171 DOI: 10.1016/j.yfrne.2006.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2006] [Accepted: 06/12/2006] [Indexed: 12/24/2022]
Abstract
We are examining the role of insulin-like growth factor-I (IGF-I) and downstream signal transduction pathways associated with growth factors (e.g., mitogen-activated protein kinase, MAPK) in estradiol and progesterone facilitation of female reproductive behavior in rats. Brain IGF-I receptor activity is required for the long-term, priming actions of estradiol on the female reproductive axis. Infusions of an IGF-I receptor antagonist during estradiol priming blocks induction of hypothalamic alpha(1B)-adrenergic receptors and luteinizing hormone surges, and attenuates lordosis behavior. Infusion of MAPK and phosphatidylinositol-3-kinase inhibitors inhibitors during estradiol priming completely blocks hormone-facilitated lordosis. Because progestin receptors (PRs) can be phosphorylated and activated by MAPKs, growth factor signaling pathways may also participate in progesterone facilitation of reproductive behaviors. Infusion of a MAPK inhibitor in estradiol-primed rats blocks progestin facilitation and sequential inhibition of lordosis and proceptive behaviors. Interference with MAPK signaling also inhibits behavioral responses to cGMP and a delta-opioid agonist, both of which can activate MAPK in some cells. Thus MAPK is involved in the facilitation of lordosis and proceptive behaviors, perhaps by phosphorylation of hypothalamic PRs.
Collapse
Affiliation(s)
- Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | | | |
Collapse
|
50
|
Korkmaz B, Ozveren E, Buharalioglu CK, Tunctan B. Extracellular Signal-Regulated Kinase (ERK1/2) Contributes to Endotoxin-Induced Hyporeactivity via Nitric Oxide and Prostacyclin Production in Rat Aorta. Pharmacology 2006; 78:123-8. [PMID: 17008773 DOI: 10.1159/000095962] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Accepted: 08/17/2006] [Indexed: 01/22/2023]
Abstract
This study was conducted to determine if mitogen-activated protein kinases (MAPKs) such as extracellular signal-regulated kinase (ERK1/2) contribute to endotoxin-induced vascular hyporeactivity via nitric oxide (NO) and/or prostacyclin (PGI(2)) production in the rat isolated thoracic aorta. Incubation of endothelium-intact rings with endotoxin (100 microg/ml) for 4 h decreased the E(max) value and increased the EC(50) value of norepinephrine. The endotoxin-induced increase in the EC(50) value of norepinephrine was decreased by phenylene-1,3-bis[ethane-2-isothiourea] dihydrobromide (1,3-PBIT), a selective inducible NO synthase inhibitor, and U0126, a selective inhibitor of ERK1/2 phosphorylation by MAPK kinase. The endotoxin-induced decrease in the E(max) value of norepinephrine was reversed by 1,3-PBIT and further decreased by U0126. 1,3-PBIT and U0126 decreased the endotoxin-induced increase in the tissue nitrite and 6-keto-PGF(1)(alpha) levels. These data suggest that events related to the activation of ERK1/2 contribute to the endotoxin-induced hyporeactivity by increasing NO and PGI(2) production.
Collapse
Affiliation(s)
- Belma Korkmaz
- Department of Pharmacology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | | | | | | |
Collapse
|