1
|
Hu Q, Qu W, Zhang T, Feng J, Dong X, Nie R, Chen J, Wang X, Peng C, Ke X. C1q/Tumor Necrosis Factor-Related Protein-9 Is a Novel Vasculoprotective Cytokine That Restores High Glucose-Suppressed Endothelial Progenitor Cell Functions by Activating the Endothelial Nitric Oxide Synthase. J Am Heart Assoc 2024; 13:e030054. [PMID: 38348774 PMCID: PMC11010095 DOI: 10.1161/jaha.123.030054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 01/10/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND This study investigated whether gCTRP9 (globular C1q/tumor necrosis factor-related protein-9) could restore high-glucose (HG)-suppressed endothelial progenitor cell (EPC) functions by activating the endothelial nitric oxide synthase (eNOS). METHODS AND RESULTS EPCs were treated with HG (25 mmol/L) and gCTRP9. Migration, adhesion, and tube formation assays were performed. Adiponectin receptor 1, adiponectin receptor 2, and N-cadherin expression and AMP-activated protein kinase, protein kinase B, and eNOS phosphorylation were measured by Western blotting. eNOS activity was determined using nitrite production measurement. In vivo reendothelialization and EPC homing assays were performed using Evans blue and immunofluorescence in mice. Treatment with gCTRP9 at physiological levels enhanced migration, adhesion, and tube formation of EPCs. gCTRP9 upregulated the phosphorylation of AMP-activated protein kinase, protein kinase B, and eNOS and increased nitrite production in a concentration-dependent manner. Exposure of EPCs to HG-attenuated EPC functions induced cellular senescence and decreased eNOS activity and nitric oxide synthesis; the effects of HG were reversed by gCTRP9. Protein kinase B knockdown inhibited eNOS phosphorylation but did not affect gCTRP9-induced AMP-activated protein kinase phosphorylation. HG impaired N-cadherin expression, but treatment with gCTRP9 restored N-cadherin expression after HG stimulation. gCTRP9 restored HG-impaired EPC functions through both adiponectin receptor 1 and N-cadherin-mediated AMP-activated protein kinase /protein kinase B/eNOS signaling. Nude mice that received EPCs treated with gCTRP9 under HG medium showed a significant enhancement of the reendothelialization capacity compared with those with EPCs incubated under HG conditions. CONCLUSIONS CTRP9 promotes EPC migration, adhesion, and tube formation and restores these functions under HG conditions through eNOS-mediated signaling mechanisms. Therefore, CTRP9 modulation could eventually be used for vascular healing after injury.
Collapse
Affiliation(s)
- Qingsong Hu
- Department of CardiologyFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Wan Qu
- Health Management CenterFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Tao Zhang
- Department of CardiologyFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Jianyi Feng
- Department of CardiologyFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Xiaobian Dong
- Department of CardiologyFirst Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Ruqiong Nie
- Department of Cardiology, Guangdong Province Key Laboratory of Arrhythmia and ElectrophysiologySun Yat‐Sen Memorial Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Junyu Chen
- Department of CardiologyFuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat‐Sen Cardiovascular Hospital)ShenzhenChina
| | - Xiaoqing Wang
- Department of CardiologyFuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat‐Sen Cardiovascular Hospital)ShenzhenChina
| | - Changnong Peng
- Department of CardiologyFuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat‐Sen Cardiovascular Hospital)ShenzhenChina
| | - Xiao Ke
- Department of CardiologyFuwai Hospital, Chinese Academy of Medical Sciences (Shenzhen Sun Yat‐Sen Cardiovascular Hospital)ShenzhenChina
| |
Collapse
|
2
|
Tripska K, Igreja Sá IC, Vasinova M, Vicen M, Havelek R, Eissazadeh S, Svobodova Z, Vitverova B, Theuer C, Bernabeu C, Nachtigal P. Monoclonal anti-endoglin antibody TRC105 (carotuximab) prevents hypercholesterolemia and hyperglycemia-induced endothelial dysfunction in human aortic endothelial cells. Front Med (Lausanne) 2022; 9:845918. [PMID: 36160139 PMCID: PMC9490272 DOI: 10.3389/fmed.2022.845918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Endoglin (Eng) is a co-receptor of the transforming growth factor β superfamily playing an important role in endothelial dysfunction. TRC105 (carotuximab) is a monoclonal antibody that blocks Eng and its downstream Smad signaling pathway. Here we have investigated for the first time the effects of TRC105 treatment on the development of endothelial dysfunction induced by 7-ketocholesterol (7K) or high glucose (HG), focusing on Eng expression, signaling, and function. In the hypercholesterolemia study, human aortic endothelial cells (HAoECs) were treated with TRC105 (300 μg/ml) for 1 h, followed by the addition of 7K (10 μg/ml) for another 12 h. In the hyperglycemia study, HAoECs were exposed to HG (45 mM) for 60 h, followed by the addition of TRC105 for another 12 h, and cells treated with 5mM glucose and 40 mM mannitol served as control. Protein levels, adhesion, and transmigration of monocytes were assessed by flow cytometry, mRNA expression was measured by qRT-PCR. 7K and HG treatment increased protein levels of NF-κB and Eng and adhesion and transmigration of monocytes through HAoECs monolayer. TRC105 pretreatment reduced the 7K- or HG-induced Eng protein levels and pSmad1/5 and pSmad2/3 signaling. Despite increased protein levels of P-selectin and VCAM-1, TRC105 mediated blockage of Eng prevented 7K- and HG-induced adhesion and transmigration of monocytes through endothelial monolayers. These results suggest that TRC105-mediated Eng blockage can counteract the hypercholesterolemia- and hyperglycemia-induced endothelial dysfunction in HAoECs, suggesting that Eng might be a potential therapeutic target in disorders associated with elevated cholesterol and glucose levels.
Collapse
Affiliation(s)
- Katarina Tripska
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Ivone Cristina Igreja Sá
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Martina Vasinova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Matej Vicen
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Radim Havelek
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Zuzana Svobodova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Barbora Vitverova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Charles Theuer
- Tracon Pharmaceuticals, Inc., San Diego, CA, United States
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czechia
- *Correspondence: Petr Nachtigal,
| |
Collapse
|
3
|
Hypotension in hereditary cardiomyopathy. Pflugers Arch 2022; 474:517-527. [PMID: 35141778 DOI: 10.1007/s00424-022-02669-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/14/2022] [Accepted: 01/22/2022] [Indexed: 12/25/2022]
Abstract
It is well accepted that hypertension may lead to the development of heart failure (HF). However, little is known about the development of hypotension that may contribute to the onset of hereditary cardiomyopathy (HCM), thus promoting heart failure and early death. The purpose of this study is to verify whether a decrease in blood pressure takes place during different phases of HCM (asymptomatic, necrosis, hypertrophy, and heart failure). Using the well-known animal model, the UM-X7.1 hamster strain of HCM (HCMH), our results showed the absence of a change in mean arterial pressure (MAP) during the asymptomatic phase preceding the development of necrosis in HCMHs when compared to age-matched normal hamster (NH). However, there was a progressive decrease in MAP that reached its lowest level during the heart failure phase. The MAP during the development of the necrosis phase of HCM was accompanied by a significant increase in the level of the sodium-hydrogen exchanger, NHE1. Treatments with the potent NHE1 inhibitor, EMD 87580 (rimeporide), did not affect MAP of NH. However, treatments with EMD 87580 during the three phases of the development of HCM significantly reversed the hypotension associated with HCM.Our results showed that the development of HCM is associated with hypotension. These results suggest that a decrease in blood pressure could be a biomarker signal for HCM leading to HF and early death. Since the blockade of NHE1 significantly but partially prevented the reduction in MAP, this suggests that other mechanisms can contribute to the development of hypotension in HCM.
Collapse
|
4
|
Huang P, Hansen JS, Saba KH, Bergman A, Negoita F, Gourdon P, Hagström-Andersson A, Lindkvist-Petersson K. Aquaglyceroporins and orthodox aquaporins in human adipocytes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2022; 1864:183795. [PMID: 34627746 DOI: 10.1016/j.bbamem.2021.183795] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 11/15/2022]
Abstract
Aquaporins play a crucial role in water homeostasis in the human body, and recently the physiological importance of aquaporins as glycerol channels have been demonstrated. The aquaglyceroporins (AQP3, AQP7, AQP9 and AQP10) represent key glycerol channels, enabling glycerol flux across the membranes of cells. Adipocytes are the major source of glycerol and during lipolysis, glycerol is released to be metabolized by other tissues through a well-orchestrated process. Here we show that both AQP3 and AQP7 bind to the lipid droplet protein perilipin 1 (PLIN1), suggesting that PLIN1 is involved in the coordination of the subcellular translocation of aquaglyceroporins in human adipocytes. Moreover, in addition to aquaglyceroporins, we discovered by transcriptome sequencing that AQP1 is expressed in human primary adipocytes. AQP1 is mainly a water channel and thus is thought to be involved in the response to hyper-osmotic stress by efflux of water during hyperglycemia. Thus, this data suggests a contribution of both orthodox aquaporin and aquaglyceroporin in human adipocytes to maintain the homeostasis of glycerol and water during fasting and feeding.
Collapse
Affiliation(s)
- Peng Huang
- Experimental Medical Science, Medical Structural Biology, BMC C13, Lund University, SE-221 84 Lund, Sweden
| | - Jesper S Hansen
- Experimental Medical Science, Medical Structural Biology, BMC C13, Lund University, SE-221 84 Lund, Sweden
| | - Karim H Saba
- Department of Laboratory Medicine, Division of Clinical Genetics, BMC C13, Lund University, 22184 SE Lund, Sweden
| | - Anna Bergman
- Department of Laboratory Medicine, Division of Clinical Genetics, BMC C13, Lund University, 22184 SE Lund, Sweden
| | - Florentina Negoita
- Experimental Medical Science, BMC C11, Lund University, SE-221 84 Lund, Sweden
| | - Pontus Gourdon
- Experimental Medical Science, Medical Structural Biology, BMC C13, Lund University, SE-221 84 Lund, Sweden
| | - Anna Hagström-Andersson
- Department of Laboratory Medicine, Division of Clinical Genetics, BMC C13, Lund University, 22184 SE Lund, Sweden
| | - Karin Lindkvist-Petersson
- Experimental Medical Science, Medical Structural Biology, BMC C13, Lund University, SE-221 84 Lund, Sweden; LINXS-Lund Institute of Advanced Neutron and X-ray Science, Scheelevägen 19, SE-223 70 Lund, Sweden.
| |
Collapse
|
5
|
Ritter M, Bresgen N, Kerschbaum HH. From Pinocytosis to Methuosis-Fluid Consumption as a Risk Factor for Cell Death. Front Cell Dev Biol 2021; 9:651982. [PMID: 34249909 PMCID: PMC8261248 DOI: 10.3389/fcell.2021.651982] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
The volumes of a cell [cell volume (CV)] and its organelles are adjusted by osmoregulatory processes. During pinocytosis, extracellular fluid volume equivalent to its CV is incorporated within an hour and membrane area equivalent to the cell's surface within 30 min. Since neither fluid uptake nor membrane consumption leads to swelling or shrinkage, cells must be equipped with potent volume regulatory mechanisms. Normally, cells respond to outwardly or inwardly directed osmotic gradients by a volume decrease and increase, respectively, i.e., they shrink or swell but then try to recover their CV. However, when a cell death (CD) pathway is triggered, CV persistently decreases in isotonic conditions in apoptosis and it increases in necrosis. One type of CD associated with cell swelling is due to a dysfunctional pinocytosis. Methuosis, a non-apoptotic CD phenotype, occurs when cells accumulate too much fluid by macropinocytosis. In contrast to functional pinocytosis, in methuosis, macropinosomes neither recycle nor fuse with lysosomes but with each other to form giant vacuoles, which finally cause rupture of the plasma membrane (PM). Understanding methuosis longs for the understanding of the ionic mechanisms of cell volume regulation (CVR) and vesicular volume regulation (VVR). In nascent macropinosomes, ion channels and transporters are derived from the PM. Along trafficking from the PM to the perinuclear area, the equipment of channels and transporters of the vesicle membrane changes by retrieval, addition, and recycling from and back to the PM, causing profound changes in vesicular ion concentrations, acidification, and-most importantly-shrinkage of the macropinosome, which is indispensable for its proper targeting and cargo processing. In this review, we discuss ion and water transport mechanisms with respect to CVR and VVR and with special emphasis on pinocytosis and methuosis. We describe various aspects of the complex mutual interplay between extracellular and intracellular ions and ion gradients, the PM and vesicular membrane, phosphoinositides, monomeric G proteins and their targets, as well as the submembranous cytoskeleton. Our aim is to highlight important cellular mechanisms, components, and processes that may lead to methuotic CD upon their derangement.
Collapse
Affiliation(s)
- Markus Ritter
- Center for Physiology, Pathophysiology and Biophysics, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
- Institute for Physiology and Pathophysiology, Paracelsus Medical University, Nuremberg, Germany
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis und Rehabilitation, Salzburg, Austria
- Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Nikolaus Bresgen
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | | |
Collapse
|
6
|
Madonna R, Doria V, Minnucci I, Pucci A, Pierdomenico DS, De Caterina R. Empagliflozin reduces the senescence of cardiac stromal cells and improves cardiac function in a murine model of diabetes. J Cell Mol Med 2020; 24:12331-12340. [PMID: 32940423 PMCID: PMC7687009 DOI: 10.1111/jcmm.15699] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/15/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022] Open
Abstract
The sodium‐glucose cotransporter 2 (SGLT2) inhibitor empagliflozin reduces heart failure in diabetes, but underlying mechanisms remain elusive. We hypothesized that empagliflozin could counteract the senescence of cardiac stromal cells (CSC), the action of which limits cardiac damage and cardiac fibrosis in diabetic‐like conditions in vitro and in vivo. CSC were isolated from murine heart biopsies (n = 5) through cardiosphere (CSp) formation and incubated for 3 or 48 hours with 5.5 mmol/L normal glucose (NG), high glucose (12‐5 and 30.5 mmol/L, HG) or a hyperosmolar control of mannitol (HM) in the presence or absence of empagliflozin 100 nmol/L. The senescent CSC status was verified by β‐gal staining and expression of the pro‐survival marker Akt (pAkt) and the pro‐inflammatory marker p38 (p‐P38). The cardiac effects of empagliflozin were also studied in vivo by echocardiography and by histology in a murine model of streptozotocin (STZ)‐induced diabetes. Compared to NG, incubations with HG and HM significantly reduced the number of CSps, increased the β‐gal‐positive CSC and P‐p38, while decreasing pAkt, all reversed by empagliflozin (P < .01). Empagliflozin also reversed cardiac dysfunction, cardiac fibrosis and cell senescence in mice with (STZ)‐induced diabetes (P < .01). Empagliflozin counteracts the pro‐senescence effect of HG and of hyperosmolar stress on CSC, and improves cardiac function via decreasing cardiac fibrosis and senescence in diabetic mice, possibly through SGLT2 off‐target effects. These effects may explain empagliflozin unexpected benefits on cardiac function in diabetic patients.
Collapse
Affiliation(s)
| | - Vanessa Doria
- Center of Aging Sciences and Translational Medicine - CESI-Met "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Ilaria Minnucci
- Center of Aging Sciences and Translational Medicine - CESI-Met "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | - Angela Pucci
- Histopathology Department, Pisa University Hospital, Pisa, Italy
| | - Donato Sante Pierdomenico
- Center of Aging Sciences and Translational Medicine - CESI-Met "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy
| | | |
Collapse
|
7
|
Madonna R, Pieragostino D, Rossi C, Confalone P, Cicalini I, Minnucci I, Zucchelli M, Del Boccio P, De Caterina R. Simulated hyperglycemia impairs insulin signaling in endothelial cells through a hyperosmolar mechanism. Vascul Pharmacol 2020; 130:106678. [DOI: 10.1016/j.vph.2020.106678] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/18/2020] [Accepted: 03/25/2020] [Indexed: 12/24/2022]
|
8
|
Amelioration of diastolic dysfunction by dapagliflozin in a non-diabetic model involves coronary endothelium. Pharmacol Res 2020; 157:104781. [PMID: 32360273 DOI: 10.1016/j.phrs.2020.104781] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 01/13/2023]
Abstract
The results of trials with sodium-glucose cotransporter 2 (SGLT2) inhibitors raised the possibility that this class of drugs provides cardiovascular benefits independently from their anti-diabetic effects, although the mechanisms are unknown. Therefore, we tested the effects of SGLT2 inhibitor dapagliflozin on the progression of experimental heart disease in a non-diabetic model of heart failure with preserved ejection fraction. Dahl salt-sensitive rats were fed a high-salt diet to induce hypertension and diastolic dysfunction and were then treated with dapagliflozin for six weeks. Dapagliflozin ameliorated diastolic function as documented by echo-Doppler and heart catheterization, while blood pressure remained markedly elevated. Chronic in vivo treatment with dapagliflozin reduced diastolic Ca2+ and Na+ overload and increased Ca2+ transient amplitude in ventricular cardiomyocytes, although no direct action of dapagliflozin on isolated cardiomyocytes was observed. Dapagliflozin reversed endothelial activation and endothelial nitric oxide synthase deficit, with reduced cardiac inflammation and consequent attenuation of pro-fibrotic signaling. The potential involvement of coronary endothelium was supported by the endothelial upregulation of Na+/H+ exchanger 1in vivo and direct effects on dapagliflozin on the activity of this exchanger in endothelial cells in vitro. In conclusions, several mechanisms may cumulatively play a significant role in the dapagliflozin-associated cardioprotection. Dapagliflozin ameliorates diastolic function and exerts a positive effect on the myocardium, possibly targeting coronary endothelium. The lower degree of endothelial dysfunction, inflammation and fibrosis translate into improved myocardial performance.
Collapse
|
9
|
Hsu CC, Chien KH, Yarmishyn AA, Buddhakosai W, Wu WJ, Lin TC, Chiou SH, Chen JT, Peng CH, Hwang DK, Chen SJ, Chang YL. Modulation of osmotic stress-induced TRPV1 expression rescues human iPSC-derived retinal ganglion cells through PKA. Stem Cell Res Ther 2019; 10:284. [PMID: 31547874 PMCID: PMC6755708 DOI: 10.1186/s13287-019-1363-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/25/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Background Transient receptor potential vanilloid 1 (TRPV1), recognized as a hyperosmolarity sensor, is a crucial ion channel involved in the pathogenesis of neural and glial signaling. Recently, TRPV1 was determined to play a role in retinal physiology and visual transmission. In this study, we sought to clarify the role of TRPV1 and the downstream pathway in the osmotic stress-related retina ganglion cell (RGC) damage. Methods First, we modified the RGC differentiation protocol to obtain a homogeneous RGC population from human induced pluripotent stem cells (hiPSCs). Subsequently, we induced high osmotic pressure in the hiPSC-derived RGCs by administering NaCl solution and observed the behavior of the TRPV1 channel and its downstream cascade. Results We obtained a purified RGC population from the heterogeneous retina cell population using our modified method. Our findings revealed that TRPV1 was activated after 24 h of NaCl treatment. Upregulation of TRPV1 was noted with autophagy and apoptosis induction. Downstream protein expression analysis indicated increased phosphorylation of CREB and downregulated brain-derived neurotrophic factor (BDNF). However, hyperosmolarity-mediated defective morphological change and apoptosis of RGCs, CREB phosphorylation, and BDNF downregulation were abrogated after concomitant treatment with the PKA inhibitor H89. Conclusion Collectively, our study results indicated that the TRPV1–PKA pathway contributed to cellular response under high levels of osmolarity stress; furthermore, the PKA inhibitor had a protective effect on RGCs exposed to this stress. Therefore, our findings may assist in the treatment of eye diseases involving RGC damage.
Collapse
Affiliation(s)
- Chih-Chien Hsu
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Ke-Hung Chien
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Aliaksandr A Yarmishyn
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Waradee Buddhakosai
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Wen-Ju Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Tai-Chi Lin
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Hwa Chiou
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Chi-Hsien Peng
- Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital and Fu-Jen Catholic University, Taipei, Taiwan
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| | - Yuh-Lih Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan. .,Department of Pharmacy, Taipei Veterans General Hospital; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
10
|
Xu YX, Huang C, Liu M, Chen N, Chen W, Yang C, Zhao Y, Li X, Duan J, Liu S, Yang S. Survivin regulated by autophagy mediates hyperglycemia-induced vascular endothelial cell dysfunction. Exp Cell Res 2018; 364:152-159. [DOI: 10.1016/j.yexcr.2018.01.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/21/2017] [Accepted: 01/28/2018] [Indexed: 12/11/2022]
|
11
|
Madonna R, Pieragostino D, Balistreri CR, Rossi C, Geng YJ, Del Boccio P, De Caterina R. Diabetic macroangiopathy: Pathogenetic insights and novel therapeutic approaches with focus on high glucose-mediated vascular damage. Vascul Pharmacol 2018; 107:S1537-1891(17)30322-1. [PMID: 29425894 DOI: 10.1016/j.vph.2018.01.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/22/2017] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
Diabetic macroangiopathy - a specific form of accelerated atherosclerosis - is characterized by intra-plaque new vessel formation due to excessive/abnormal neovasculogenesis and angiogenesis, increased vascular permeability of the capillary vessels, and tissue edema, resulting in frequent atherosclerotic plaque hemorrhage and plaque rupture. Mechanisms that may explain the premature and rapidly progressive nature of atherosclerosis in diabetes are multiple, and to a large extent still unclear. However, mechanisms related to hyperglycemia certainly play an important role. These include a dysregulated vascular regeneration. In addition, oxidative and hyperosmolar stresses, as well as the activation of inflammatory pathways triggered by a dysregulated activation of membrane channel proteins aquaporins, have been recognized as key events. Here, we review recent knowledge of cellular and molecular pathways of macrovascular disease related to hyperglycemia in diabetes. We also here highlight how new insights into pathogenic mechanisms of vascular damage in diabetes may indicate new targets for prevention and treatment.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy; Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Damiana Pieragostino
- Analitical Biochemistry and Proteomics Unit Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy
| | - Carmela Rita Balistreri
- Department of Patho-biology and Medical Biotechnologies, University of Palermo, Palermo, Italy
| | - Claudia Rossi
- Analitical Biochemistry and Proteomics Unit Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy
| | - Yong-Jian Geng
- Center for Cardiovascular Biology and Atherosclerosis Research, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Piero Del Boccio
- Analitical Biochemistry and Proteomics Unit Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy
| | - Raffaele De Caterina
- Center of Aging Sciences and Translational Medicine - CESI-MeT, Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti, Italy.
| |
Collapse
|
12
|
Sada K, Nishikawa T, Kukidome D, Yoshinaga T, Kajihara N, Sonoda K, Senokuchi T, Motoshima H, Matsumura T, Araki E. Hyperglycemia Induces Cellular Hypoxia through Production of Mitochondrial ROS Followed by Suppression of Aquaporin-1. PLoS One 2016; 11:e0158619. [PMID: 27383386 PMCID: PMC4934928 DOI: 10.1371/journal.pone.0158619] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 06/20/2016] [Indexed: 01/02/2023] Open
Abstract
We previously proposed that hyperglycemia-induced mitochondrial reactive oxygen species (mtROS) generation is a key event in the development of diabetic complications. Interestingly, some common aspects exist between hyperglycemia and hypoxia-induced phenomena. Thus, hyperglycemia may induce cellular hypoxia, and this phenomenon may also be involved in the pathogenesis of diabetic complications. In endothelial cells (ECs), cellular hypoxia increased after incubation with high glucose (HG). A similar phenomenon was observed in glomeruli of diabetic mice. HG-induced cellular hypoxia was suppressed by mitochondria blockades or manganese superoxide dismutase (MnSOD) overexpression, which is a specific SOD for mtROS. Overexpression of MnSOD also increased the expression of aquaporin-1 (AQP1), a water and oxygen channel. AQP1 overexpression in ECs suppressed hyperglycemia-induced cellular hypoxia, endothelin-1 and fibronectin overproduction, and apoptosis. Therefore, hyperglycemia-induced cellular hypoxia and mtROS generation may promote hyperglycemic damage in a coordinated manner.
Collapse
Affiliation(s)
- Kiminori Sada
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Nishikawa
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Molecular Diabetology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- * E-mail:
| | - Daisuke Kukidome
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoaki Yoshinaga
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Nobuhiro Kajihara
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuhiro Sonoda
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takafumi Senokuchi
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Department of Molecular Diabetology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Motoshima
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Matsumura
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
13
|
Madonna R, Giovannelli G, Confalone P, Renna FV, Geng YJ, De Caterina R. High glucose-induced hyperosmolarity contributes to COX-2 expression and angiogenesis: implications for diabetic retinopathy. Cardiovasc Diabetol 2016; 15:18. [PMID: 26822858 PMCID: PMC4731895 DOI: 10.1186/s12933-016-0342-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/22/2016] [Indexed: 12/26/2022] Open
Abstract
Background We tested the hypothesis that glucose-induced hyperosmolarity, occurring in diabetic hyperglycemia, promotes retinal angiogenesis, and that interference with osmolarity signaling ameliorates excessive angiogenesis and retinopathy in vitro and in vivo. Methods and Results We incubated human aortic (HAECs) and dermal microvascular endothelial cells (HMVECs) with glucose or mannitol for 24 h and tested them for protein levels and in vitro angiogenesis. We used the Ins2 Akita mice as a model of type 1 diabetes to test the in vivo relevance of in vitro observations. Compared to incubations with normal (5 mmol/L) glucose concentrations, cells exposed to both high glucose and high mannitol (at 30.5 or 50.5 mmol/L) increased expression of the water channel aquaporin-1 (AQP1) and cyclooxygenase (COX)-2. This was preceded by increased activity of the osmolarity-sensitive transcription factor Tonicity enhancer binding protein (TonEBP), and enhanced endothelial migration and tubulization in Matrigel, reverted by treatment with AQP1 and TonEBP siRNA. Retinas of Ins2 Akita mice showed increased levels of AQP1 and COX-2, as well as angiogenesis, all reverted by AQP1 siRNA intravitreal injections. Conclusions Glucose-related hyperosmolarity seems to be able to promote angiogenesis and retinopathy through activation of TonEBP and possibly increasing expression of AQP1 and COX-2. Osmolarity signaling may be a target for therapy.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Laboratory of Experimental Cardiology, Center of Excellence on Aging, Institute of Cardiology, "G. d'Annunzio" University, C/o Ospedale SS. Annunziata, Via dei Vestini, 31, 66013, Chieti, Italy. .,The University of Texas Health Science Center at Houston and the Texas Heart Institute, Houston, TX, USA.
| | - Gaia Giovannelli
- Department of Neurosciences and Imaging, "G. d'Annunzio" University, Chieti, Italy.
| | - Pamela Confalone
- Laboratory of Experimental Cardiology, Center of Excellence on Aging, Institute of Cardiology, "G. d'Annunzio" University, C/o Ospedale SS. Annunziata, Via dei Vestini, 31, 66013, Chieti, Italy.
| | - Francesca Vera Renna
- Laboratory of Experimental Cardiology, Center of Excellence on Aging, Institute of Cardiology, "G. d'Annunzio" University, C/o Ospedale SS. Annunziata, Via dei Vestini, 31, 66013, Chieti, Italy.
| | - Yong-Jian Geng
- The University of Texas Health Science Center at Houston and the Texas Heart Institute, Houston, TX, USA.
| | - Raffaele De Caterina
- Laboratory of Experimental Cardiology, Center of Excellence on Aging, Institute of Cardiology, "G. d'Annunzio" University, C/o Ospedale SS. Annunziata, Via dei Vestini, 31, 66013, Chieti, Italy.
| |
Collapse
|
14
|
The ethanol extract of Zingiber zerumbet rhizomes mitigates vascular lesions in the diabetic retina. Vascul Pharmacol 2015; 76:18-27. [PMID: 26319672 DOI: 10.1016/j.vph.2015.08.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/29/2015] [Accepted: 08/23/2015] [Indexed: 11/19/2022]
Abstract
Diabetic retinopathy (DR) is a common diabetic eye disease which is well-known as the result of microvascular retinal changes. Although the ethanol extract from Zingiber zerumbet (L.) Smith rhizome (EEZZR) has been indicated to ameliorate hyperglycemia in diabetes, its protective effect on DR remains unclear. The aim of this study was to determine the effects of EEZZR on DR in streptozotocin (STZ) diabetic rats. Diabetic rats were treated orally with EEZZR (200, 300 mg/kg per day) or calcium dobesilate (CD; 500 mg/kg per day) for 12 weeks. EEZZR displayed similar characteristics to CD in reducing blood-retinal barrier permeability in diabetic rats. Retinal histopathological observation showed that retinal vessels were decreased in EEZZR-treated diabetic rats. EEZZR decreased the increased retinal expression of vascular endothelial growth factor (VEGF) and upregulate the expressions of renal pigment epithelium-derived factor (PEDF) in diabetic rats. Retinal mRNA expression of tumor necrosis factor-α, interleukin (IL)-1, IL-6, monocyte chemotactic proteins-1, intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 were all decreased in EEZZR-treated diabetic rats. Moreover, EEZZR could attenuate phosphorylation of nuclear factor Kappa B (NF-κB) p65 and extracellular signal-regulated kinase (ERK)1/2 as well as inhibit the nuclear translocation of pNF-κB p65 induced by diabetes. In conclusion, restoring the balance between stimulators and inhibitors of angiogenesis may be associated with the protective effect of EEZZR on DR. In addition, EEZZR can ameliorate retinal inflammation via transrepression of NF-κB and inhibition of ERK1/2 signaling pathway.
Collapse
|
15
|
del Nogal M, Troyano N, Calleros L, Griera M, Rodriguez-Puyol M, Rodriguez-Puyol D, Ruiz-Torres MP. Hyperosmolarity induced by high glucose promotes senescence in human glomerular mesangial cells. Int J Biochem Cell Biol 2014; 54:98-110. [DOI: 10.1016/j.biocel.2014.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/07/2014] [Accepted: 07/10/2014] [Indexed: 02/06/2023]
|
16
|
Transplantation of Mesenchymal Cells Improves Peripheral Limb Ischemia in Diabetic Rats. Mol Biotechnol 2014; 56:438-48. [DOI: 10.1007/s12033-014-9735-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
17
|
Madonna R, De Caterina R. Sodium-hydrogen exchangers (NHE) in human cardiovascular diseases: interfering strategies and their therapeutic applications. Vascul Pharmacol 2013; 59:127-30. [PMID: 24140414 DOI: 10.1016/j.vph.2013.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 10/06/2013] [Indexed: 11/29/2022]
Abstract
Sodium-hydrogen exchangers (NHE) are among the main regulators of cell volume and intracellular concentration of hydrogen and sodium ions. By indirectly affecting sodium/calcium exchange across the plasma membrane, NHE can also influence the intracellular concentration of calcium. Excess activation of NHE or inappropriate sodium extrusion due to failure of ATP-dependent Na(+)/K(+) transport system can be deleterious during cardiac or peripheral organ ischemia. Besides being responsible for the regulation of intracellular pH and sodium-calcium inward currents, NHE isoform 1 (NHE-1), which is predominantly expressed in the cardiovascular system, influences the tone of the vessel wall in response to a variety of stimuli, including hypertonic stress. Because of the extensive involvement of NHE-1 in cardiac myocyte contracture and necrosis, stunning, reperfusion arrhythmias, as well as hypertension and myocardial diseases such as diabetic cardiomyopathy, efforts have been made in developing inhibitors of this transporter. We here review the biology and regulation of NHE, focusing on current knowledge of the role of NHE-1 as a potential target in the development of novel compounds that could play a role in cardiovascular homeostasis, both in physiological and pathological conditions.
Collapse
|
18
|
Madonna R, Jiang J, Geng YJ. Attenuated expression of gelsolin in association with induction of aquaporin-1 and nitric oxide synthase in dysfunctional hearts of aging mice exposed to endotoxin. Int J Immunopathol Pharmacol 2013; 25:911-22. [PMID: 23298482 DOI: 10.1177/039463201202500409] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis triggered by endotoxinemia may impair cardiac function. A decline in tolerance to septic shock occurs with aging. This study addressed the hypothesis that aging negatively impairs expression of gelsolin, and axerts the regulatory effects on the water channel protein aquaporin-1 (AQP-1) and endotoxin-inducible nitric oxide synthase (iNOS). We explored whether the age-related gene changes are associated with the cardiac dysfunction induced by endotoxic stress exposure. Male mice at young (3-month) and old (12-month) ages received intraperitoneal injections of saline or lipopolysaccharide (LPS, 30mg/Kg). Cardiac performance and morphology were analyzed by echocardiography at baseline and 2 and 24 h after injection. At the end of treatment, the animals were sacrificed, and cardiac tissues were collected for assessing expression of gelsolin, AQP-1, iNOS, and transcription-3 (STAT3). LPS administration led to a decreased contractility while increasing cardiac dimensions in both young and old mice. LPS also markedly induced expression of gelsolin in both animal groups. However, compared to young mice, old mice showed compromised induction of gelsolin and cardiac performance in response to endotoxin. Meanwhile, the LPS-exposed old animals exhibited higher levels of AQP-1, iNOS, and phosphorylated STAT3. Gelsolin-null mice had increased expression of glycosylated AQP-1 and STAT3 phosphorylation as well as cardiac dysfunction. Thus, endotoxin administration induces expression of gelsolin, AQP-1 and pro-inflammatory genes, such as iNOS. Our data suggest that changed expression of gelsolin, AQP-1 and iNOS may contribute to dysfunction of hearts in aged subjects with septic endotoxinemia.
Collapse
Affiliation(s)
- R Madonna
- The University of Texas Health Science Center, Houston, TX, USA
| | | | | |
Collapse
|
19
|
Madonna R, Cevik C, Nasser M. Electrical plasticity and cardioprotection in myocardial ischemia--role of selective sodium channel blockers. Clin Cardiol 2013; 36:255-61. [PMID: 23529949 DOI: 10.1002/clc.22113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 02/11/2013] [Indexed: 11/08/2022] Open
Abstract
The concept of electrical protection of the ischemic myocardium is in constant evolution and has recently been supported by experimental and clinical studies. Historically, antiplatelet agents, angiotensin-converting enzyme inhibitors, β-blockers, and statins have been all proposed as drugs conferring anti-ischemic cardioprotection. This was supported by the evidence consistently indicating that all these drugs were capable of reducing mortality and the risk of repeat myocardial infarction. The electrical plasticity paradigm is, however, a novel concept that depicts the benefits of improved sodium channel blockade with drugs such as ranolazine and cariporide. Although it has been hypothesized that the protective role of ranolazine depends on decreased fatty acid β-oxidation affecting preconditioning, we speculate against such a hypothesis, because inhibition of β-oxidation requires higher concentrations of the drug, above the therapeutic range. Rather, we discuss the key role of calcium overload reduction through inhibition of the late sodium current (I(Na)). Mechanisms driving cardioprotection involve the block of a cascade of complex ionic exchanges that can result in intracellular acidosis, excess cytosolic calcium, myocardial cellular dysfunction, and eventually cell injury and death. In this review we discuss the studies that demonstrate how electrical plasticity through sodium channel blockers can promote cardioprotection against ischemia in coronary heart disease.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Texas Heart Institute and St. Luke's Episcopal Hospital, Department of adult cardiiology, Houston, Texas, USA.
| | | | | |
Collapse
|
20
|
Sasahara T, Yayama K, Matsuzaki T, Tsutsui M, Okamoto H. Na(+)/H(+) exchanger inhibitor induces vasorelaxation through nitric oxide production in endothelial cells via intracellular acidification-associated Ca2(+) mobilization. Vascul Pharmacol 2012; 58:319-25. [PMID: 23268360 DOI: 10.1016/j.vph.2012.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 11/12/2012] [Accepted: 11/24/2012] [Indexed: 10/27/2022]
Abstract
The objective of this study was to determine the mechanism by which Na(+)/H(+) exchanger (NHE) inhibitors induce vasodilatation. The NHE inhibitors, 5-(N,N-dimethyl)-amiloride (DMA), cariporide, and amiloride, evoked endothelium-dependent relaxation in rat aortas with ED50 values of 16, 89, and 148μM, respectively, and these effects were abolished by treatment with N(G)-nitro-l-arginine methyl ester (L-NAME). The relaxation effects induced by DMA and cariporide were strongly attenuated in aortas of the endothelial NO synthase (eNOS)-deficient mice, as compared to the effects in wild-type mice. The DMA-induced relaxation in rat aorta was attenuated by a calmodulin (CaM) inhibitor, calmidazolium, and a soluble guanylyl cyclase inhibitor, [1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, but was not affected by a phosphoinositide 3-kinase inhibitor, wortmannin. Immunoblots for endothelial eNOS on immunoprecipitated CaM complexes showed that DMA enhanced the association of eNOS with CaM in rat aortas. Both DMA and cariporide induced the reduction of intracellular pH (pHi) in bovine aortic endothelial cells (BAECs), which was accompanied by a sustained elevation of cytosolic Ca(2+) ([Ca(2+)]i). This DMA-induced rise of [Ca(2+)]i was not affected by removing external Ca(2+) from the buffer, but was abolished in thapsigargin-pretreated BAECs. These results suggest that lowering of pHi by NHE inhibitors in endothelial cells induces the mobilization of Ca(2+) from the thapsigargin-sensitive stores of endoplasmic reticulum, which in turn stimulates NO production via the CaM-dependent activation of eNOS.
Collapse
Affiliation(s)
- Tomoya Sasahara
- Laboratory of Cardiovascular Pharmacology, Department of Biopharmaceutical Sciences, Kobe Gakuin University, Minatojima 1-1-3, Chuo-ku, Kobe 650-8586, Japan
| | | | | | | | | |
Collapse
|
21
|
Delli Pizzi S, Madonna R, Caulo M, Romani GL, De Caterina R, Tartaro A. MR angiography, MR imaging and proton MR spectroscopy in-vivo assessment of skeletal muscle ischemia in diabetic rats. PLoS One 2012; 7:e44752. [PMID: 23028603 PMCID: PMC3448608 DOI: 10.1371/journal.pone.0044752] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 08/06/2012] [Indexed: 02/04/2023] Open
Abstract
To prospectively evaluate the feasibility of using magnetic resonance (MR) techniques for in-vivo assessing a rat diabetic model of limb ischemia. Unilateral hind limb ischemia was induced by ligation of the iliac-femoral artery in male streptozotocin-treated and non-diabetic control rats. Four weeks after ligation, rats underwent MR Angiography (MRA), T1-weighted and Short Time Inversion Recovery (STIR) sequences and muscle Proton MR Spectroscopy (1H-MRS) on both hind limbs. After MR examinations, immunoblotting and immunofluorescence analysis were performed. MRA showed a signal void due to flow discontinuation distal to the artery ligation. T1-weighted and STIR images showed, respectively, the presence of tissue swelling (p = 0.018 for non-diabetic; p = 0.027 for diabetic rats) and signal hyperintensity in tissue affected by occlusion. Mean total creatine/water for the occluded limb was significantly lower than for the non-occluded limbs in both non-diabetic (5.46×10−4 vs 1.14×10−3, p = 0.028) and diabetic rats (1.37×10−4 vs 1.10×10−3; p = 0.018). MR Imaging and 1H-MRS changes were more pronounced in diabetic than in non-diabetic occluded limbs (p = 0.032). MR findings were confirmed by using histological findings. Combined MR techniques can be used to demonstrate the presence of structural and metabolic changes produced by iliac-femoral artery occlusion in rat diabetic model of limb ischemia.
Collapse
Affiliation(s)
- Stefano Delli Pizzi
- Department of Neuroscience and Imaging, Institute for Advanced Biomedical Technologies, “G. d'Annunzio University” Foundation, Chieti, Italy
| | - Rosalinda Madonna
- Department of Neuroscience and Imaging, Cardiology Division of Center of Excellence on Aging, University “G. d'Annunzio”, Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience and Imaging, Institute for Advanced Biomedical Technologies, “G. d'Annunzio University” Foundation, Chieti, Italy
| | - Gian Luca Romani
- Department of Neuroscience and Imaging, Institute for Advanced Biomedical Technologies, “G. d'Annunzio University” Foundation, Chieti, Italy
| | - Raffaele De Caterina
- Department of Neuroscience and Imaging, Cardiology Division of Center of Excellence on Aging, University “G. d'Annunzio”, Chieti, Italy
| | - Armando Tartaro
- Department of Neuroscience and Imaging, Institute for Advanced Biomedical Technologies, “G. d'Annunzio University” Foundation, Chieti, Italy
- * E-mail:
| |
Collapse
|
22
|
Tetè S, Tripodi D, Rosati M, Conti F, Maccauro G, Saggini A, Salini V, Cianchetti E, Caraffa A, Antinolfi P, Toniato E, Castellani ML, Pandolfi F, Frydas S, Conti P, Theoharides TC. Endothelial cells, cholesterol, cytokines, and aging. Int J Immunopathol Pharmacol 2012; 25:355-63. [PMID: 22697067 DOI: 10.1177/039463201202500205] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
It has been reported that high levels of cholesterol and triglycerides are associated with increased risk of developing atherosclerosis and shorter life. In fact, vascular endothelial dysfunction occurs during the human aging process. Accumulation of lipids in vascular endothelium activates leukocytes to produce cytokines and chemokines which recruit macrophages. On the other hand, macrophages augment inflammatory response and secrete vascular endothelial growth factor, a key cytokine that mediates angiogenesis and inflammatory response. In addition, hyperlipidaemia is one of the main risk factors for aging, hypertension and diabetes. Here, we review the interrelationship between endothelial cells, high level of cholesterol, and aging.
Collapse
|
23
|
Ohanian J, Forman SP, Katzenberg G, Ohanian V. Endothelin-1 Stimulates Small Artery VCAM-1 Expression through p38MAPK-Dependent Neutral Sphingomyelinase. J Vasc Res 2012; 49:353-62. [DOI: 10.1159/000336649] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Accepted: 01/10/2012] [Indexed: 01/21/2023] Open
|
24
|
Salini V, Saggini A, Maccauro G, Caraffa A, Shaik-Dasthagirisaheb Y, Conti P. Inflammatory Markers: Serum Amyloid A, Fibrinogen and C-Reactive Protein — A Revisited Study. EUR J INFLAMM 2011. [DOI: 10.1177/1721727x1100900202] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The acute phase response is the part of the innate defence system of an animal against trauma, inflammation or infection. During this response, there is increased production and release of certain plasma proteins known as acute phase proteins, which include C-reactive protein (CRP), serum amyloid A (SAA) and fibrinogen (Fg). CRP consists of five identical subunits of 206 amino acids with a molecular weight of approximately 23 kDa. There is strong evidence from numerous studies that CRP is a predictor of inflammation. The acute-phase protein serum amyloid A (SAA) is a clinically useful marker of inflammation. SAA plays not only an important role in the development of AA amyloidosis (an important complication of rheumatoid arthritis) but also interacts with events closely involved in the metabolic syndrome as a high- and low-grade inflammatory modulator. Fibrinogen (Fg) is a high molecular weight plasma adhesion protein and is a biomarker of inflammation. It is synthesized and assembled in hepatocytes and fibroblasts and when secreted into the circulation, its plasma half-life ranges from 3 to 4 days. Several cytokines, are involved in the induction of acute phase protein synthesis, but the mutual importance of these cytokines seems to be cell-type specific and to vary in various experimental settings. Here we revisited the classic acute phase proteins SAA, C-Reactive protein and fibrinogen in their role in inflammation and their interrelationship with some cytokines.
Collapse
Affiliation(s)
- V. Salini
- Orthopaedic Division University of Chieti-Pescara, Medical School, Chieti, Italy
| | - A. Saggini
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - G. Maccauro
- Department of Orthopaedics, Catholic University of Rome, Rome, Italy
| | - A. Caraffa
- Orthopaedics Division, University of Perugia, Perugia, Italy
| | | | - P. Conti
- Immunology Division, University of Chieti-Pescara, Medical School, Chieti, Italy
| |
Collapse
|
25
|
Madonna R, De Caterina R. Cellular and molecular mechanisms of vascular injury in diabetes--part II: cellular mechanisms and therapeutic targets. Vascul Pharmacol 2011; 54:75-9. [PMID: 21453785 DOI: 10.1016/j.vph.2011.03.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 03/19/2011] [Indexed: 11/29/2022]
Abstract
Although the mechanisms by which insulin-resistance and hyperglycemia lead to cardiovascular disease are still incompletely understood, all mechanisms apparently converge on the vessel wall and the endothelium as a common disease target. Endothelial cells play a crucial role in vascular homeostasis, providing a functional barrier and modulating several signals involved in vasomotion, as well as antiplatelet, anti-inflammatory, anti-proliferative, and anti-oxidant properties of the vessel wall. Endothelial cell dysfunction occurs early in diabetes and insulin resistance states. Since atherosclerosis may result from an imbalance between the magnitude of vascular injury and the capacity of repair, a role has been recently postulated for a defective mobilization of vascular progenitors, including endothelial progenitor cells, in the pathogenesis of vascular disease. Here we summarize the evidence for such an occurrence. We also here highlight how new insights into pathways of vascular damage in diabetes may indicate new targets for preventive and treatment strategies.
Collapse
|