1
|
Lima AFR, Rodrigues D, Machado MR, Oliveira-Neto JT, Bressan AFM, Pedersoli CA, Alves JV, Silva-Neto JA, Barros PR, Dias TB, Garcia LV, Bruder-Nascimento A, Bruder-Nascimento T, Carneiro FS, Leiria LOS, Tostes RC, Costa RM. Endothelin-1 down-regulates nuclear factor erythroid 2-related factor-2 and contributes to perivascular adipose tissue dysfunction in obesity. Clin Sci (Lond) 2024; 138:1071-1087. [PMID: 39136472 DOI: 10.1042/cs20240624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024]
Abstract
Perivascular adipose tissue (PVAT) negatively regulates vascular muscle contraction. However, in the context of obesity, the PVAT releases vasoconstrictor substances that detrimentally affect vascular function. A pivotal player in this scenario is the peptide endothelin-1 (ET-1), which induces oxidative stress and disrupts vascular function. The present study postulates that obesity augments ET-1 production in the PVAT, decreases the function of the nuclear factor erythroid 2-related factor-2 (Nrf2) transcription factor, further increasing reactive oxygen species (ROS) generation, culminating in PVAT dysfunction. Male C57BL/6 mice were fed either a standard or a high-fat diet for 16 weeks. Mice were also treated with saline or a daily dose of 100 mg·kg-1 of the ETA and ETB receptor antagonist Bosentan, for 7 days. Vascular function was evaluated in thoracic aortic rings, with and without PVAT. Mechanistic studies utilized PVAT from all groups and cultured WT-1 mouse brown adipocytes. PVAT from obese mice exhibited increased ET-1 production, increased ECE1 and ETA gene expression, loss of the anticontractile effect, as well as increased ROS production, decreased Nrf2 activity, and downregulated expression of Nrf2-targeted antioxidant genes. PVAT of obese mice also exhibited increased expression of Tyr216-phosphorylated-GSK3β and KEAP1, but not BACH1 - negative Nrf2 regulators. Bosentan treatment reversed all these effects. Similarly, ET-1 increased ROS generation and decreased Nrf2 activity in brown adipocytes, events mitigated by BQ123 (ETA receptor antagonist). These findings place ET-1 as a major contributor to PVAT dysfunction in obesity and highlight that pharmacological control of ET-1 effects restores PVAT's cardiovascular protective role.
Collapse
Affiliation(s)
- Anna Flavia R Lima
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Daniel Rodrigues
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Mirele R Machado
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - José Teles Oliveira-Neto
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Alecsander F M Bressan
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Carina A Pedersoli
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Juliano V Alves
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Júlio A Silva-Neto
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Paula R Barros
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Thiago B Dias
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Luis V Garcia
- Department of Biomechanics, Medicine and Locomotive Apparatus Rehabilitation, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | | | - Fernando S Carneiro
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Luiz Osório S Leiria
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Rafael M Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, U.S.A
- Institute of Health Sciences, Federal University of Jatai, Jatai, GO, Brazil
| |
Collapse
|
2
|
Edgerton-Fulton M, Ergul A. Vascular contributions to cognitive impairment/dementia in diabetes: role of endothelial cells and pericytes. Am J Physiol Cell Physiol 2022; 323:C1177-C1189. [PMID: 36036445 PMCID: PMC9576164 DOI: 10.1152/ajpcell.00072.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/19/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022]
Abstract
Vascular contributions to cognitive impairment/dementia (VCID) are a leading cause of dementia, a known neurodegenerative disorder characterized by progressive cognitive decline. Although diabetes increases the risks of stroke and the development of cerebrovascular disease, the cellular and vascular mechanisms that lead to VCID in diabetes are yet to be determined. A growing body of research has identified that cerebrovascular cells within the neurovascular complex display an array of cellular responses that impact their survival and reparative properties, which plays a significant role in VCID development. Specifically, endothelial cells and pericytes are the primary cell types that have gained much attention in dementia-related studies due to their molecular and phenotypic heterogeneity. In this review, we will discuss the various morphological subclasses of endothelial cells and pericytes as well as their relative distribution throughout the cerebrovasculature. Furthermore, the use of diabetic and stroke animal models in preclinical studies has provided more insight into the impact of sex differences on cerebral vascularization in progressive VCID. Understanding how cellular responses and sex differences contribute to endothelial cell and pericyte survival and function will set the stage for the development of potential preventive therapies for dementia-related disorders in diabetes.
Collapse
Affiliation(s)
- Mia Edgerton-Fulton
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Adviye Ergul
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
3
|
Guo Y, Zuo W, Yin L, Gu T, Wang S, Fang Z, Wang B, Dong H, Hou W, Zuo Z, Deng J. Pioglitazone Attenuates Ischemic Stroke Aggravation By Blocking PPARγ Reduction and Inhibiting Chronic Inflammation in Diabetic Mice. Eur J Neurosci 2022; 56:4948-4961. [PMID: 35945686 DOI: 10.1111/ejn.15789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/03/2022] [Indexed: 11/28/2022]
Abstract
Diabetes can cause vascular remodeling and is associated with worse outcome after ischemic stroke. Pioglitazone is a commonly used anti-diabetic agent. However, it is not known whether pioglitazone use before ischemia could reduce brain ischemic injury. Pioglitazone was administered to 5-week-old db+ or db/db mice. Cerebral vascular remodeling was examined at the age of 9 weeks. Expression of peroxisome proliferator-activated receptor-γ (PPARγ), p-PPARγ (S112 and S273), nucleotide-binding domain (NOD)-like receptor protein 3 (Nlrp3), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) was evaluated in the somatosensory cortex of mice. Neurological outcome was evaluated 24 h after brain ischemia. Results showed that early pioglitazone treatment provided a long-lasting effect of euglycemia but enhanced hyperlipidemia in the db/db mice. Diabetic mice exhibited increased vascular tortuosity, narrower middle cerebral artery (MCA) width and IgG leakage in the brain. These changes were blocked by early pioglitazone treatment. In diabetic animals, PPARγ expression was reduced and p-PPARγ at S273 but not S112, Nlrp3, IL-1β and TNF-α were increased in the somatosensory cortex. PPARγ decrease and Nlrp3 increase were mainly in the neurons of the diabetic brain, which was reversed by early pioglitazone treatment. Pioglitazone attenuated the aggravated neurological outcome after stroke in diabetic mice. But this protective effect was abolished through restoring cerebral inflammation by intracerebroventricular administration of IL-1β and TNF-α in pioglitazone treated diabetic mice before MCAO. In summary, early pioglitazone treatment attenuates cerebral vascular remodeling and ischemic brain injury possibly via blocking chronic neuroinflammation in the db/db mice.
Collapse
Affiliation(s)
- Yaru Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lu Yin
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tingting Gu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zongping Fang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bairen Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Anesthesiology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
4
|
Impact of diabetes and ischemic stroke on the cerebrovasculature: A female perspective. Neurobiol Dis 2022; 167:105667. [PMID: 35227927 PMCID: PMC9615543 DOI: 10.1016/j.nbd.2022.105667] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/25/2022] [Accepted: 02/17/2022] [Indexed: 01/16/2023] Open
Abstract
There is a very complex interaction between the brain and the cerebral vasculature to meet the metabolic demands of the brain for proper function. Preservation of vascular networks and cerebrovascular function ultimately plays a key role in this intricate communication within the brain in health and disease. Experimental evidence showed that diabetes not only affects the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression, but also alters cerebrovascular function resulting in compromised myogenic reactivity and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain can rapidly occur. When an ischemic insult is superimposed on this pathology, not only is the neurovascular injury greater, but repair mechanisms fail, resulting in greater physical and cognitive deficits. While clinically it is known that women suffer disproportionately from diabetes as well as ischemic stroke and post-stroke cognitive impairment, the cerebrovascular architecture, patho/physiology, as well as cerebrovascular contributions to stroke recovery in female and diabetic animal models are inadequately studied and highlighted in this review.
Collapse
|
5
|
Derella CC, Blanks AM, Nguyen A, Looney J, Tucker MA, Jeong J, Rodriguez-Miguelez P, Thomas J, Lyon M, Pollock DM, Harris RA. Dual endothelin receptor antagonism increases resting energy expenditure in people with increased adiposity. Am J Physiol Endocrinol Metab 2022; 322:E508-E516. [PMID: 35373585 PMCID: PMC9126219 DOI: 10.1152/ajpendo.00349.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 01/18/2023]
Abstract
Increased adiposity is associated with dysregulation of the endothelin system, both of which increase the risk of cardiovascular disease (CVD). Preclinical data indicate that endothelin dysregulation also reduces resting energy expenditure (REE). The objective was to test the hypothesis that endothelin receptor antagonism will increase REE in people with obesity compared with healthy weight individuals. Using a double blind, placebo-controlled, crossover design, 32 participants [healthy weight (HW): n = 16, BMI: 21.3 ± 2.8 kg/m2, age: 26 ± 7 yr and overweight/obese (OB): n = 16, BMI: 33.5 ± 9.5 kg/m2, age: 31 ± 6 yr] were randomized to receive either 125 mg of bosentan (ETA/B antagonism) or placebo twice per day for 3 days. Breath-by-breath gas exchange data were collected and REE was assessed by indirect calorimetry. Venous blood samples were analyzed for concentrations of endothelin-1 (ET-1). Treatment with bosentan increased plasma ET-1 in both OB and HW groups. Within the OB group, the changes in absolute REE (PLA: -77.6 ± 127.6 vs. BOS: 72.2 ± 146.6 kcal/day; P = 0.046). The change in REE was not different following either treatment in the HW group. Overall, absolute plasma concentrations of ET-1 following treatment with bosentan were significantly associated with kcal/day of fat (r = 0.488, P = 0.005), percentage of fat utilization (r = 0.415, P = 0.020), and inversely associated with the percentage of carbohydrates (r = -0.419, P = 0.019), and respiratory exchange ratio (r = -0.407, P = 0.023). Taken together, these results suggest that modulation of the endothelin system may represent a novel therapeutic approach to increase both resting metabolism and caloric expenditure, and reduce CVD risk in people with increased adiposity.NEW & NOTEWORTHY Findings from our current translational investigation demonstrate that dual endothelin A/B receptor antagonism increases total REE in overweight/obese individuals. These results suggest that modulation of the endothelin system may represent a novel therapeutic target to increase both resting metabolism and caloric expenditure, enhance weight loss, and reduce CVD risk in seemingly healthy individuals with elevated adiposity.
Collapse
Affiliation(s)
- Cassandra C Derella
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Anson M Blanks
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Andy Nguyen
- Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jacob Looney
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Matthew A Tucker
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Jinhee Jeong
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Paula Rodriguez-Miguelez
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Jeffrey Thomas
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
| | - Matthew Lyon
- Medical College of Georgia, Augusta University, Augusta, Georgia
| | - David M Pollock
- Cardio-Renal Physiology and Medicine Section, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ryan A Harris
- Department of Medicine, Georgia Prevention Institute, Augusta University, Augusta, Georgia
- Sport and Exercise Science Research Institute, Ulster University, Jordanstown, United Kingdom
| |
Collapse
|
6
|
Chaumais MC, Djessas MRA, Thuillet R, Cumont A, Tu L, Hebert G, Gaignard P, Huertas A, Savale L, Humbert M, Guignabert C. Additive protective effects of sacubitril/valsartan and bosentan on vascular remodelling in experimental pulmonary hypertension. Cardiovasc Res 2021; 117:1391-1401. [PMID: 32653925 DOI: 10.1093/cvr/cvaa200] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/03/2020] [Accepted: 07/03/2020] [Indexed: 01/26/2023] Open
Abstract
AIMS Although right ventricular (RV) function is an important determinant of morbidity and mortality in patients with pulmonary arterial hypertension (PAH), there is no treatment targeting directly the RV. We evaluate the efficacy of sacubitril/valsartan (LCZ 696) as add-on therapy to bosentan in rats with severe pulmonary hypertension (PH). METHODS AND RESULTS Combination therapy of LCZ 696 and bosentan has additive vascular protective effects against the pulmonary vascular remodelling and PH in two preclinical models of severe PH. Compared with monotherapy, co-treatment of LCZ 696 (30 or 68 mg/kg/day for 2 weeks, per os) and bosentan (100 mg/kg/day for 2 weeks, per os) started 7 days after monocrotaline (MCT) injection substantially reduces pulmonary pressures, vascular remodelling, and RV hypertrophy and fibrosis in rats. Consistent with these observations, co-treatment of rats with established PH induced by sugen/hypoxia (SuHx) with LCZ 696 (30 mg/kg/day for 3 weeks, per os) and bosentan (100 mg/kg/day for 3 weeks, per os) started 5 weeks after Sugen injection partially attenuate total pulmonary vascular resistance and cardiovascular structures. We also obtained evidence showing that LCZ 696 has anti-proliferative effect on cultured human pulmonary artery smooth muscle cells derived from patients with idiopathic PAH, an effect that is more pronounced in presence of bosentan. Finally, we found that the plasma levels of atrial natriuretic peptide (ANP) and cyclic guanosine monophosphate (cGMP) are higher in rats co-treated with LCZ 696 (30 mg/kg/day) and bosentan (100 mg/kg/day) than in MCT and SuHx rats treated with vehicle. CONCLUSION Dual therapy with LCZ 696 plus bosentan proved significantly superior beneficial effect to LCZ 696 or bosentan alone on vascular remodelling and severity of experimental PH.
Collapse
MESH Headings
- Aminobutyrates/pharmacology
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Atrial Natriuretic Factor/blood
- Biphenyl Compounds/pharmacology
- Bosentan/pharmacology
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic GMP/blood
- Disease Models, Animal
- Disease Progression
- Drug Combinations
- Drug Therapy, Combination
- Endothelin Receptor Antagonists/pharmacology
- Familial Primary Pulmonary Hypertension/drug therapy
- Familial Primary Pulmonary Hypertension/metabolism
- Familial Primary Pulmonary Hypertension/physiopathology
- Humans
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neprilysin/antagonists & inhibitors
- Protease Inhibitors/pharmacology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Rats, Wistar
- Valsartan/pharmacology
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Marie-Camille Chaumais
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Pharmacie, 92290, Châtenay-Malabry, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pharmacie, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Mohamed Reda Amar Djessas
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Amélie Cumont
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Ly Tu
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Guillaume Hebert
- Hôpital Marie Lannelongue, Service pharmacie, 92350, Le Plessis-Robinson, France
| | - Pauline Gaignard
- Université Paris-Saclay, Faculté de Pharmacie, 92290, Châtenay-Malabry, France
- AP-HP, Laboratoire de biochimie, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Alice Huertas
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Laurent Savale
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, 94270, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM, UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| |
Collapse
|
7
|
Guo Z, Wu X, Fan W. Clarifying the effects of diabetes on the cerebral circulation: Implications for stroke recovery and beyond. Brain Res Bull 2021; 171:67-74. [PMID: 33662495 DOI: 10.1016/j.brainresbull.2021.02.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
Given the sheer increased number of victims per year and the availability of only one effective treatment, acute ischemic stroke (AIS) remains to be one of the most under-treated serious diseases. Diabetes not only increases the incidence of ischemic stroke, but amplifies the ischemic damage, upon which if patients with diabetes suffer from stroke, he/she will confront increased risks of long-term functional deficits. The grim reality makes it a pressing need to intensify efforts at the basic science level to understand how diabetes impairs stroke recovery. This review retrospects the clinical and experimental studies in order to elucidate the detrimental effect of diabetes on cerebrovascular circulation including the major arteries/arterioles, collateral circulation, and neovascularization to shed light on further exploration of novel strategies for cerebral circulation protection before and after AIS in patients with diabetes.
Collapse
Affiliation(s)
- Zhihui Guo
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Xuqing Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| | - Wei Fan
- Department of Neurology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
8
|
Ali EMT, Abdallah HI, El-Sayed SM. Histomorphological, VEGF and TGF-β immunoexpression changes in the diabetic rats' ovary and the potential amelioration following treatment with metformin and insulin. J Mol Histol 2020; 51:287-305. [PMID: 32399705 DOI: 10.1007/s10735-020-09880-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus (DM) affects the ovary by reducing the number and diameters of ovarian follicles and increasing atretic follicles. Follicular growth and diameters depend on VEGF production. Hyperglycemia causes ovarian stromal and follicular degeneration then fibrosis by activating TGF-β. Insulin and metformin promote development of ovarian follicles and reduce atretic follicles. Therefore, the present study investigates the ovarian VEGF and TGF-β immune-expression and its variations in diabetic, insulin and metformin-treated rats. Forty adult female albino rats were divided equally into four groups: control, diabetic (STZ-induced diabetes), diabetic metformin-treated group (100 mg/kg/day orally/eight weeks) and diabetic insulin-treated group (5 U insulin /day). Ovarian sections were stained with hematoxylin and eosin, Masson's trichrome, immunohistochemistry for VEGF and TGF-β. The diabetic group showed noticeable atrophic and degenerative changes in cortex and medulla as well as increased density and distribution of the collagenous fibers. The number and diameter of primary, secondary and tertiary follicles were decreased. However, the number of atretic follicles and corpus luteum was increased. Significant decrease in the surface area percentage of VEGF immuno-expression and significant increase in TGF-β immuno-expression surface area percentage were detected. By treating animals with metformin and insulin, there was restoration of the ovarian histological structure more or less as in control. DM negatively affects the histological and morphometric parameters of ovaries. Furthermore, insulin showed more beneficial effects than metformin in hindering these complications by modifying the expression of VEGF and TGF-β.
Collapse
Affiliation(s)
- Eyad M T Ali
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia. .,Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Hesham I Abdallah
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia.,Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sayed M El-Sayed
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia.,Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Abdul Y, Jamil S, He L, Li W, Ergul A. Endothelin-1 (ET-1) promotes a proinflammatory microglia phenotype in diabetic conditions. Can J Physiol Pharmacol 2020; 98:596-603. [PMID: 32119570 DOI: 10.1139/cjpp-2019-0679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Diabetes increases the risk and severity of cognitive impairment, especially after ischemic stroke. It is also known that the activation of the endothelin (ET) system is associated with cognitive impairment and microglia around the periinfarct area produce ET-1. However, little is known about the effect of ET-1 on microglial polarization, especially under diabetic conditions. We hypothesized that (i) ET-1 activates microglia to the proinflammatory M-1-like phenotype and (ii) hypoxia/ lipopolysaccharide (LPS) activates the microglial ET system and promotes microglial activation towards the M-1 phenotype in diabetic conditions. Microglial cells (C8B4) cultured under normal-glucose (25 mmol/L) conditions and diabetes-mimicking high-glucose (50 mmol/L) conditions for 48 h were stimulated with ET-1, cobalt chloride (200 μmol/L), or LPS (100 ng/mL) for 24 h. PPET-1, ET receptor subtypes, and M1/M2 marker gene mRNA expression were measured by RT-PCR. Secreted ET-1 was measured by ELISA. A high dose of ET-1 (1 μmol/L) increases the mRNA levels of ET receptors and activates the microglia towards the M1 phenotype. Hypoxia or LPS activates the ET system in microglial cells and shifts the microglia towards the M1 phenotype in diabetic conditions. These in vitro observations warrant further investigation into the role of ET-1-mediated activation of proinflammatory microglia in post-stroke cognitive impairment in diabetes.
Collapse
Affiliation(s)
- Yasir Abdul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave., Charleston, SC 29425-2503, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Sarah Jamil
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave., Charleston, SC 29425-2503, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Lianying He
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave., Charleston, SC 29425-2503, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Weiguo Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave., Charleston, SC 29425-2503, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 171 Ashley Ave., Charleston, SC 29425-2503, USA.,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| |
Collapse
|
10
|
Halvorson BD, Whitehead SN, McGuire JJ, Wiseman RW, Frisbee JC. Endothelium-dependent impairments to cerebral vascular reactivity with type 2 diabetes mellitus in the Goto-Kakizaki rat. Am J Physiol Regul Integr Comp Physiol 2019; 317:R149-R159. [PMID: 31091154 DOI: 10.1152/ajpregu.00088.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent pathology associated with elevated cerebrovascular disease risk. We determined wall mechanics and vascular reactivity in ex vivo middle cerebral arteries (MCA) from male Goto-Kakizaki rats (GK; ~17 wk old) versus control Wistar Kyoto rats (WKY) to test the hypothesis that the diabetic environment in GK, in the absence of obesity and other comorbidities, leads to endothelial dysfunction and impaired vascular tone regulation. Dilation of MCA following challenge with acetylcholine and hypoxia was blunted in MCA from GK versus WKY, due to lower nitric oxide bioavailability and altered arachidonic acid metabolism, whereas myogenic activation and constrictor responses to serotonin were unchanged. MCA wall distensibility and cross-sectional area were not different between GK and WKY, suggesting that wall mechanics were unchanged at this age, supported by the determination that MCA dilation to sodium nitroprusside was also intact. With the use of ex vivo aortic rings as a bioassay, altered vascular reactivity determined in MCA was paralleled by relaxation responses in artery segments from GK, whereas measurements of vasoactive metabolite production indicated a loss of nitric oxide and prostacyclin bioavailability and an increased thromboxane A2 production with both methacholine challenge and hypoxia. These results suggest that endothelium-dependent dilator reactivity of MCA in GK is impaired with T2DM, and that this impairment is associated with the genesis of a prooxidant/pro-inflammatory condition with diabetes mellitus. The restriction of vascular impairments to endothelial function only, at this age and development, provide insight into the severity of multimorbid conditions of which T2DM is only one constituent.
Collapse
Affiliation(s)
- Brayden D Halvorson
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario , London, Ontario , Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario , London, Ontario , Canada
| | - John J McGuire
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario , London, Ontario , Canada
| | - Robert W Wiseman
- Departments of Physiology and Radiology, Michigan State University , East Lansing, Michigan
| | - Jefferson C Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario , London, Ontario , Canada
| |
Collapse
|
11
|
Fouda AY, Fagan SC, Ergul A. Brain Vasculature and Cognition. Arterioscler Thromb Vasc Biol 2019; 39:593-602. [PMID: 30816798 PMCID: PMC6540805 DOI: 10.1161/atvbaha.118.311906] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
There is a complex interaction between the brain and the cerebral vasculature to meet the metabolic demands of the brain for proper function. Preservation of cerebrovascular function and integrity has a central role in this sophisticated communication within the brain, and any derangements can have deleterious acute and chronic consequences. In almost all forms of cognitive impairment, from mild to Alzheimer disease, there are changes in cerebrovascular function and structure leading to decreased cerebral blood flow, which may initiate or worsen cognitive impairment. In this focused review, we discuss the contribution of 2 major vasoactive pathways to cerebrovascular dysfunction and cognitive impairment in an effort to identify early intervention strategies.
Collapse
Affiliation(s)
- Abdelrahman Y. Fouda
- Vascular Biology Center, Augusta University, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Susan C. Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Adviye Ergul
- Ralph Johnson Veterans Administration Medical Center, Medical University of South Carolina, Charleston, SC
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
12
|
Coucha M, Barrett AC, Elgebaly M, Ergul A, Abdelsaid M. Inhibition of Ephrin-B2 in brain pericytes decreases cerebral pathological neovascularization in diabetic rats. PLoS One 2019; 14:e0210523. [PMID: 30620753 PMCID: PMC6324788 DOI: 10.1371/journal.pone.0210523] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/22/2018] [Indexed: 12/20/2022] Open
Abstract
We have previously shown that diabetes causes dysfunctional cerebral neovascularization that increases the risk for cerebrovascular disorders such as stroke and cognitive impairment. Pericytes (PCs) play a pivotal role in the angiogenic process through their interaction with the endothelial cells (EC). Yet, the role of PCs in dysfunctional cerebral neovascularization in diabetes is unclear. In the present study, we tested the hypothesis that the increased proangiogenic Ephrin-B2 signaling in PCs contributes to the dysfunctional cerebral neovascularization in diabetes. Type-II diabetes was induced by a combination of high fat diet and low dose streptozotocin injection in male Wistar rats. Selective in vivo Ephrin-B2 silencing in brain PCs was achieved using the stereotactic injection of adeno-associated virus (AAV) with NG2-promoter that expresses Ephrin-B2 shRNA. Neovascularization was assessed using vascular fluorescent dye stain. Novel object recognition (NOR) test was used to determine cognitive functions. Human brain microvascular pericytes HBMVPCs were grown in high glucose 25 mM and palmitate 200 uM (HG/Pal) to mimic diabetic conditions. Scratch migration and tube formation assays were conducted to evaluate PC/EC interaction and angiogenic functions in PC/EC co-culture. Diabetes increased the expression of Ephrin-B2 in the cerebrovasculature and pericytes. Concomitant increases in cerebral neovascularization parameters including vascular density, tortuosity and branching density in diabetic rats were accompanied by deterioration of cognitive function. Inhibition of Ephrin-B2 expression in PCs significantly restored cerebral vascularization and improved cognitive functions. HG/Pal increased PC/EC angiogenic properties in co-culture. Silencing Ephrin-B2 in PCs significantly reduced PC migration and PC/EC co-culture angiogenic properties. This study emphasizes the significant contribution of PCs to the pathological neovascularization in diabetes. Our findings introduce Ephrin-B2 signaling as a promising therapeutic target to improve cerebrovascular integrity in diabetes.
Collapse
Affiliation(s)
- Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Amy C. Barrett
- Biomedical Sciences Department, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Mostafa Elgebaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, Florida, United States of America
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Mohammed Abdelsaid
- Biomedical Sciences Department, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| |
Collapse
|
13
|
Li W, Abdul Y, Ward R, Ergul A. Endothelin and diabetic complications: a brain-centric view. Physiol Res 2018; 67:S83-S94. [PMID: 29947530 DOI: 10.33549/physiolres.933833] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The global epidemic of diabetes is of significant concern. Diabetes associated vascular disease signifies the principal cause of morbidity and mortality in diabetic patients. It is also the most rapidly increasing risk factor for cognitive impairment, a silent disease that causes loss of creativity, productivity, and quality of life. Small vessel disease in the cerebral vasculature plays a major role in the pathogenesis of cognitive impairment in diabetes. Endothelin system, including endothelin-1 (ET-1) and the receptors (ET(A) and ET(B)), is a likely candidate that may be involved in many aspects of the diabetes cerebrovascular disease. In this review, we took a brain-centric approach and discussed the role of the ET system in cerebrovascular and cognitive dysfunction in diabetes.
Collapse
Affiliation(s)
- W Li
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA, Department of Physiology, Augusta University, Augusta, Georgia, USA.
| | | | | | | |
Collapse
|
14
|
ARFIAN N, KUSUMA MHH, ANGGOROWATI N, NUGROHO DB, JEFFILANO A, SUZUKI Y, IKEDA K, EMOTO N. Vitamin D Upregulates Endothelin-1, ETBR, eNOS mRNA Expression and Attenuates Vascular Remodelling and Ischemia in Kidney Fibrosis Model in Mice Vitamin D Upregulates Endothelin-1, ETBR, eNOS mRNA Expression and Attenuates Vascular Remodelling and Ischemia in Kidney Fibrosis Model in Mice. Physiol Res 2018; 67:S137-S147. [DOI: 10.33549/physiolres.933823] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We examined the upregulation of ET-1/ETBR/eNOS signaling in renoprotective effect of vitamin D in kidney fibrosis model in mice using unilateral ureteral obstruction (UUO). One group was treated with intraperitoneal injection of 0.125 mg/kg of Calcitriol (UUO+VD). Vascular remodeling was quantified based on lumen area and lumen/wall area ratio (LWAR) of intrarenal arteries using Sirius Red staining. ET-1, ETBR, eNOS, CD31 and VEGF mRNA expressions were quantified using qRT-PCR. Focusing on endothelin-1 (ET-1) signaling in endothelial cells (EC), siRNA of ET-1 was performed in human umbilical vein endothelial cells (HUVEC) for reducing ET-1 expression. Then HUVECs were treated with and without 100 nM Calcitriol treatment in hypoxic and normoxic conditions to elucidate ET-1/eNOS signaling. Our in vivo study revealed vascular remodeling and renal ischemia attenuation after Calcitriol treatment. Vascular remodeling was attenuated in the UUO+VD group as shown by increasing lumen areas and LWAR in intrarenal arteries. These findings were associated with significant higher CD31 and VEGF mRNA expression compared to the UUO group. Vitamin D treatment also increased ET-1, ETBR and eNOS mRNA expressions. Our in vitro study demonstrated Calcitriol induced ET-1 and eNOS mRNA expressions upregulation in HUVEC under normoxic and hypoxic condition. Meanwhile, siRNA for ET-1 inhibited the upregulation of eNOS mRNA expression after Calcitriol treatment. Vitamin D ameliorates kidney fibrosis through attenuating vascular remodeling and ischemia with upregulating ET-1/ETBR and eNOS expression.
Collapse
Affiliation(s)
- N. ARFIAN
- Department of Anatomy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Fu X, Pan Y, Cao Q, Li B, Wang S, Du H, Duan N, Li X. Metformin restores electrophysiology of small conductance calcium-activated potassium channels in the atrium of GK diabetic rats. BMC Cardiovasc Disord 2018; 18:63. [PMID: 29636010 PMCID: PMC5894224 DOI: 10.1186/s12872-018-0805-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/04/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Small conductance calcium-activated potassium channels (SK channels) play a critical role in action potential repolarization in cardiomyocytes. Recently, the potential anti-arrhythmic effect of metformin in diabetic patients has been recognized, yet the underlying mechanism remains elusive. METHODS Diabetic Goto-Kakizaki (GK) rats were untreated or treated with metformin (300 mg/kg/day) for 12 weeks, and age-matched Wistar rats were used as control (n = 6 per group). Electrocardiography, Hematoxylin-eosin staining and Masson's trichome staining were performed to assess cardiac function, histology and fibrosis. The expression levels of the SK channels in the myocardium were determined by real-time PCR and Western blotting. The electrophysiology of the SK channels in the cardiomyocytes isolated from the three groups of rats was examined by patch clamp assay, with specific blockade of the SK channels with apamin. RESULTS Metformin treatment significantly reduced cardiac fibrosis and alleviated arrhythmia in the diabetic rats. In the atrial myocytes from control, GK and metformin-treated GK rats, the expression of KCa2.2 (SK2 channel) was down-regulated and the expression of KCa2.3 (SK3 channel) was up-regulated in the atrium of GK rats as compared with that of control rats, and metformin reversed diabetes-induced alterations in atrial SK channel expression. Moreover, patch clamp assay revealed that the SK current was markedly reduced and the action potential duration was prolonged in GK atrial myocytes, and the SK channel function was partially restored in the atrial myocytes from metformin-treated GK rats. CONCLUSIONS Our data suggests an involvement of the SK channels in the development of arrhythmia under diabetic conditions, and supports a potential beneficial effect of metformin on atrial electrophysiology.
Collapse
Affiliation(s)
- Xi Fu
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Qian Cao
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Bin Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Shuo Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Hongjiao Du
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Na Duan
- Department of Cardiology, The People's Hospital of Liaoning Province, Shenyang, 110016, People's Republic of China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
16
|
Coucha M, Abdelsaid M, Ward R, Abdul Y, Ergul A. Impact of Metabolic Diseases on Cerebral Circulation: Structural and Functional Consequences. Compr Physiol 2018; 8:773-799. [PMID: 29687902 DOI: 10.1002/cphy.c170019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolic diseases including obesity, insulin resistance, and diabetes have profound effects on cerebral circulation. These diseases not only affect the architecture of cerebral blood arteries causing adverse remodeling, pathological neovascularization, and vasoregression but also alter the physiology of blood vessels resulting in compromised myogenic reactivity, neurovascular uncoupling, and endothelial dysfunction. Coupled with the disruption of blood brain barrier (BBB) integrity, changes in blood flow and microbleeds into the brain rapidly occur. This overview is organized into sections describing cerebrovascular architecture, physiology, and BBB in these diseases. In each section, we review these properties starting with larger arteries moving into smaller vessels. Where information is available, we review in the order of obesity, insulin resistance, and diabetes. We also tried to include information on biological variables such as the sex of the animal models noted since most of the information summarized was obtained using male animals. © 2018 American Physiological Society. Compr Physiol 8:773-799, 2018.
Collapse
Affiliation(s)
- Maha Coucha
- South University, School of Pharmacy, Savannah, Georgia, USA
| | | | - Rebecca Ward
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yasir Abdul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, USA.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
17
|
Di Pietro M, Pascuali N, Parborell F, Abramovich D. Ovarian angiogenesis in polycystic ovary syndrome. Reproduction 2018; 155:R199-R209. [PMID: 29386378 DOI: 10.1530/rep-17-0597] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most prevalent endocrine pathology among women in reproductive age. Its main symptoms are oligo or amenorrhea, hyperandrogenism and the presence of ovarian cysts. It is also associated with infertility, obesity and insulin resistance. Mainly due to its heterogeneity, PCOS treatments are directed to manage its symptoms and to prevent associated diseases. The correct formation and regression of blood vessels during each ovarian cycle is indispensable for proper follicular development, ovulation and corpus luteum formation. The importance of these processes opened a new and promising field: ovarian angiogenesis. Vascular alterations characterize numerous pathologies, either with increased, decreased or abnormal angiogenesis. In the last years, several anomalies of ovarian angiogenesis have been described in women with PCOS. Therefore, it has been suggested that these alterations may be associated with the decreased - or lack of - ovulation rates and for the formation of cysts in the PCOS ovaries. Restoration of a proper vessel formation in the ovaries may lead to improved follicular development and ovulation in these patients. In the present review, we attempt to summarize the alterations in ovarian angiogenesis that have been described in women with PCOS. We also discuss the therapeutic approaches aimed to correct these alterations and their beneficial effects on the treatment of infertility in PCOS.
Collapse
Affiliation(s)
- Mariana Di Pietro
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME-CONICET)Buenos Aires, Argentina
| |
Collapse
|
18
|
Lin Y, Zhao Y, Liu E. High glucose upregulates endothelin type B receptors in vascular smooth muscle cells via the downregulation of Sirt1. Int J Mol Med 2017; 41:439-445. [PMID: 29115394 DOI: 10.3892/ijmm.2017.3242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 10/17/2017] [Indexed: 11/06/2022] Open
Abstract
Silent information regulator family protein 1 (Sirt1) has recently gained attention for its protective effects against diabetic and cardiovascular diseases (CVDs). Vascular smooth muscle endothelin type B (ETB) receptors are involved in the pathogenesis of CVDs and diabetes. The aim of present study was to explore whether Sirt1 is involved in high glucose (HG)-mediated regulation of ETB receptors in rat superior mesenteric arteries (SMA). The rat SMA segments were cultured in the presence and absence of HG with or without the activator of Sirt1 and specific inhibitor for the extracellular signal-regulated protein kinase 1/2 (ERK1/2) for 24 h. Following organ culture, the contractile responses to sarafotoxin 6c were studied using a sensitive myograph, and the ETB receptor protein expression level was determined using western blotting. The results demonstrated that HG induced upregulation of ETB receptor expression and increased receptor-mediated vasoconstriction in SMA. Resveratrol (Res; a Sirt1 activator) concentration-dependently inhibited the HG-induced upregulation of ETB receptor expression and receptor-mediated vasoconstriction. Additionally, these effects could also be abolished by an inhibitor of the ERK1/2 signaling pathway. Furthermore, upregulation of ERK1/2 phosphorylation induced by HG was inhibited by Res. In conclusion, HG upregulated ETB receptors by downregulating Sirt1 and subsequently activating the ERK1/2 signaling pathways in the organ culture SMA.
Collapse
Affiliation(s)
- Yan Lin
- Xi'an Jiaotong University Cardiovascular Research Center, Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Zhao
- Department of Pathophysiology, School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Enqi Liu
- Xi'an Jiaotong University Cardiovascular Research Center, Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
19
|
He J, Yi B, Chen Y, Huang Q, Wang H, Lu K, Fu W. The ET-1-mediated carbonylation and degradation of ANXA1 induce inflammatory phenotype and proliferation of pulmonary artery smooth muscle cells in HPS. PLoS One 2017; 12:e0175443. [PMID: 28414743 PMCID: PMC5393570 DOI: 10.1371/journal.pone.0175443] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/27/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatopulmonary syndrome (HPS) is a serious complication of advanced liver disease, which markedly increases mortality. Pulmonary vascular remodelling (PVR) induced by circulating mediators plays an important role in the pathogenesis of HPS, while the underlying mechanism remains undefined. In the present study, we reported that endothelin-1 (ET-1) is up-regulated and annexin A1(ANXA1) is down-regulated in HPS rat, and ET-1 decreases the ANXA1 expression in a dose-dependent manner in rat pulmonary arterial smooth muscle cells (PASMCs). Then, we showed that ANXA1 can decrease nuclear p-ERK1/2 accumulation and decrease the cyclin D1 expression, thus resulting in the subsequent inhibition of PASMCs proliferation. As previously reported, we confirmed that ET-1 decreases the ANXA1 protein levels by the carbonylation and degradation of ANXA1. In conclusion, our research links the signaling cascade of ET1-ANXA1-cell proliferation to a potential therapeutic strategy for blocking IPS-associated PVR.
Collapse
Affiliation(s)
- Jing He
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Bin Yi
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Yang Chen
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
| | - Qing Huang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Huan Wang
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Kaizhi Lu
- Department of Anesthesia, Southwest Hospital, The Third Military Medical University, Chongqing, China
- * E-mail: (KL); (WF)
| | - Weiling Fu
- Department of Laboratory Medicine, Southwest Hospital, Third Military Medical University, Chongqing, China
- * E-mail: (KL); (WF)
| |
Collapse
|
20
|
Abdelsaid M, Coucha M, Hafez S, Yasir A, Johnson MH, Ergul A. Enhanced VEGF signalling mediates cerebral neovascularisation via downregulation of guidance protein ROBO4 in a rat model of diabetes. Diabetologia 2017; 60:740-750. [PMID: 28116460 PMCID: PMC5342922 DOI: 10.1007/s00125-017-4214-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/31/2016] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS Diabetes promotes cerebral neovascularisation via increased vascular endothelial growth factor (VEGF) angiogenic signalling. Roundabout-4 (ROBO4) protein is an endogenous inhibitor of VEGF signalling that stabilises the vasculature. Yet, how diabetes affects ROBO4 function remains unknown. We hypothesised that increased VEGF signalling in diabetes decreases ROBO4 expression and function via binding of ROBO4 with VEGF-activated β3 integrin and that restoration of ROBO4 expression prevents/repairs cerebral neovascularisation in diabetes. METHODS ROBO4 protein expression in a rat model of type 2 diabetes (Goto-Kakizaki [GK] rats) was examined by western blotting and immunohistochemistry. ROBO4 was locally overexpressed in the brain and in primary brain microvascular endothelial cells (BMVECs). GK rats were treated with SKLB1002, a selective VEGF receptor-2 (VEGFR-2) antagonist. Cerebrovascular neovascularisation indices were determined using a FITC vascular space-filling model. Immunoprecipitation was used to determine ROBO4-β3 integrin interaction. RESULTS ROBO4 expression was significantly decreased in the cerebral vasculature as well as in BMVECs in diabetes (p < 0.05). Silencing Robo4 increased the angiogenic properties of control BMVECs (p < 0.05). In vivo and in vitro overexpression of ROBO4 inhibited VEGF-induced angiogenic signalling and increased vessel maturation. Inhibition of VEGF signalling using SKLB1002 increased ROBO4 expression (p < 0.05) and reduced neovascularisation indices (p < 0.05). Furthermore, SKLB1002 significantly decreased ROBO4-β3 integrin interaction in diabetes (p < 0.05). CONCLUSIONS/INTERPRETATION Our study identifies the restoration of ROBO4 and inhibition of VEGF signalling as treatment strategies for diabetes-induced cerebral neovascularisation.
Collapse
Affiliation(s)
- Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA.
- Department of Physiology, Augusta University, 1120 15th Street CA-3135, Augusta, GA, 30912, USA.
| | - Maha Coucha
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-3135, Augusta, GA, 30912, USA
| | - Sherif Hafez
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-3135, Augusta, GA, 30912, USA
| | - Abdul Yasir
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-3135, Augusta, GA, 30912, USA
| | | | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA, USA
- Department of Physiology, Augusta University, 1120 15th Street CA-3135, Augusta, GA, 30912, USA
| |
Collapse
|
21
|
Fouda MA, Abdel-Rahman AA. Endothelin Confers Protection against High Glucose-Induced Neurotoxicity via Alleviation of Oxidative Stress. J Pharmacol Exp Ther 2017; 361:130-139. [PMID: 28179472 PMCID: PMC5363775 DOI: 10.1124/jpet.116.238659] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/06/2017] [Indexed: 11/22/2022] Open
Abstract
Recent findings linked the inhibition in the neuromodulator peptide endothelin-1 (ET-1) level to the high glucose-evoked neurotoxicity. However, definitive neuroprotective role for ET-1 and the major neuronal ET (ET-3) against high glucose-evoked toxicity and the implicated neurochemical responses triggered by their ET-A and ET-B receptors remain unknown. Here, we tested the hypothesis that ET-B activation alleviates high glucose-evoked oxidative stress and cell death. High glucose (100 mM for 48 hours)-evoked cell death was associated with elevation in reactive oxygen species, inhibition of catalase activity, and a paradoxical upregulation of hemeoxygenase-1 expression along with ET-A and ET-B receptors were downregulated and upregulated, respectively. ET-1 or ET-3, in concentrations that had no effect on PC12 cell viability in normal glucose medium, alleviated all high glucose-evoked neurochemical responses, except for the reduction in ET-A receptor expression. Prior (4 hours) incubation with a selective ET-A (BQ123) or ET-B (BQ788) receptor blocker abrogated the neuroprotection conferred by ET-1 or ET-3. However, the ET-B receptor played a greater role because BQ788 abrogated the favorable ET-1- or ET-3-mediated reversal of the ERK1/2 phosphorylation and the inhibition in catalase activity caused by high glucose. These findings suggest that endothelin exerts ET-B receptor-dependent favorable redox and neuroprotective effects against high glucose-evoked oxidative damage and neurotoxicity.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Pharmacology, Brody School of Medicine, East Carolina University, North Carolina
| | - Abdel A Abdel-Rahman
- Department of Pharmacology, Brody School of Medicine, East Carolina University, North Carolina
| |
Collapse
|
22
|
Singh M, Prakash A. Possible role of endothelin receptor against hyperhomocysteinemia and β-amyloid induced AD type of vascular dementia in rats. Brain Res Bull 2017; 133:31-41. [PMID: 28274813 DOI: 10.1016/j.brainresbull.2017.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 01/25/2017] [Accepted: 02/28/2017] [Indexed: 11/16/2022]
Abstract
Vascular dementia (VaD) is considered as the second commonest form of dementia after Alzheimer's disease (AD). The study was designed to investigate the effect of endothelin receptor against β-amyloid induced AD type of vascular dementia. This disease was induced by combine administration of single ICV (intracerebroventricle) infusion of β-amyloid (Aβ) once and chronic oral administration of l-Methionine for 21 days. Bosentan (dual endothelin receptor antagonist) was administered for 21 days. Behavioral alterations were observed during different time interval of the study. Animals were killed immediately following the last behavior session. Oxidative parameters, acetylcholinesterase activity, neuro-inflammatory markers, amyloid beta levels were determined in hippocampus and cortex while serum homocysteine, serum nitrite carotid artery superoxide anion level were also determined. Endothelial function was measured on isolated carotid artery using myograph instrument. Aβ+l-Methionine showed more significant development of cognitive and vascular endothelial deficits, manifested in terms of increase in serum homocysteine level, endothelial dysfunction, impairment of learning and memory, enhanced brain acetylcholinesterase activity, marked mito-oxidative damage in rats. We have observed that l-Methionine and combination of Aβ+l-Methionine significantly enhanced Aβ level both in cortex as well as hippocampus. Treatment of bosentan attenuated Aβ+l-Methionine induced impairment of learning and memory, enhanced Aβ level, mitochondrial and endothelial dysfunction. The results of present study concluded that bosentan offers protection against β-amyloid-induced vascular dementia in rats. Endothelin receptor may be considered as a potential pharmacological target for the management of AD type of vascular dementia.
Collapse
Affiliation(s)
- Major Singh
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga 142-001, Punjab, India
| | - Atish Prakash
- Department of Pharmacology, ISF College of Pharmacy, Ghal Kalan, Moga 142-001, Punjab, India; Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Ghasemian E, Motaghian P, Vatanara A. D-optimal Design for Preparation and Optimization of Fast Dissolving Bosentan Nanosuspension. Adv Pharm Bull 2016; 6:211-8. [PMID: 27478783 DOI: 10.15171/apb.2016.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 05/07/2016] [Accepted: 05/08/2016] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Bosentan is a drug currently taken orally for the treatment of pulmonary arterial hypertension. However, the water solubility of bosentan is very low, resulting in low bioavailability. The aim of this study was preparation and optimization of bosentan nanosuspension to improve solubility and dissolution rate. METHODS The different formulations designed by Design Expert® software. Nanosuspensions were prepared using precipitation method and the effects of stabilizer type and content and drug content on the particle size, polydispersity (PDI) and yield of nanosuspensions were investigated. RESULTS Particle size, PDI and yield of the optimal nanosuspension formulation were 200.9 nm, 0.24 and 99.6%, respectively. Scanning electron microscopy (SEM) results showed spherical morphology for bosentan nanoparticles. Thermal analysis indicated that there was a partial crystalline structure and change in the pholymorphism of bosentan in the nanoparticles. In addition, reduction of particle size, significantly increased in vitro dissolution rate of the drug. CONCLUSION Optimization by design expert software was shown to be a successful method for optimization and prediction of responses by less than 10% error and formulation with 15.8 mg span 85 as an internal stabilizer and 45 mg drug content were introduced as the optimum formulation. The solubility of bosentan in the optimal formulation was 6.9 times higher than coarse bosentan and could be suggested as promising drug delivery systems for improving the dissolution rate and possibly the pharmacokinetic of bosentan.
Collapse
Affiliation(s)
- Elham Ghasemian
- Pharmaceutics Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Motaghian
- Pharmaceutics Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Vatanara
- Pharmaceutics Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Hardigan T, Yasir A, Abdelsaid M, Coucha M, El-Shaffey S, Li W, Johnson MH, Ergul A. Linagliptin treatment improves cerebrovascular function and remodeling and restores reduced cerebral perfusion in Type 2 diabetes. Am J Physiol Regul Integr Comp Physiol 2016; 311:R466-77. [PMID: 27357799 DOI: 10.1152/ajpregu.00057.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
The antihyperglycemic agent linagliptin, a dipeptidyl peptidase-4 (DPP-IV) inhibitor, has been shown to reduce inflammation and improve endothelial cell function. In this study, we hypothesized that DPP-IV inhibition with linagliptin would improve impaired cerebral perfusion in diabetic rats, as well as improve insulin-induced cerebrovascular relaxation and reverse pathological cerebrovascular remodeling. We further postulated that these changes would lead to a subsequent improvement of cognitive function. Male Type-2 diabetic and nondiabetic Goto-Kakizaki rats were treated with linagliptin for 4 wk, and blood glucose and DPP-IV plasma levels were assessed. Cerebral perfusion was assessed after treatment using laser-Doppler imaging, and dose response to insulin (10(-13) M-10(-6) M) in middle cerebral arteries was tested on a pressurized arteriograph. The impact of DPP-IV inhibition on diabetic cerebrovascular remodeling was assessed over a physiologically relevant pressure range, and changes in short-term hippocampus-dependent learning were observed using a novel object recognition test. Linagliptin lowered DPP-IV activity but did not change blood glucose or insulin levels in diabetes. Insulin-mediated vascular relaxation and cerebral perfusion were improved in the diabetic rats with linagliptin treatment. Indices of diabetic vascular remodeling, such as increased cross-sectional area, media thickness, and wall-to-lumen ratio, were also ameliorated; however, improvements in short-term hippocampal-dependent learning were not observed. The present study provides evidence that linagliptin treatment improves cerebrovascular dysfunction and remodeling in a Type 2 model of diabetes independent of glycemic control. This has important implications in diabetic patients who are predisposed to the development of cerebrovascular complications, such as stroke and cognitive impairment.
Collapse
Affiliation(s)
- Trevor Hardigan
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Abdul Yasir
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Maha Coucha
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Sally El-Shaffey
- Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Weiguo Li
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| | - Maribeth H Johnson
- Department of Biostatistics, The Graduate School at Augusta University, Augusta, Georgia
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta, Georgia; and
| |
Collapse
|
25
|
Yuan W, Li Y, Wang J, Li J, Gou S, Fu P. Endothelin-receptor antagonists for diabetic nephropathy: A meta-analysis. Nephrology (Carlton) 2016; 20:459-66. [PMID: 25753148 DOI: 10.1111/nep.12442] [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] [Accepted: 01/06/2015] [Indexed: 02/05/2023]
Abstract
AIM Endothelin-receptor antagonists may be a novel therapeutic strategy for diabetic nephropathy, but their use remains controversial. This meta-analysis seeks to evaluate the effectiveness and safety of endothelin-receptor antagonists for patients with diabetic nephropathy. METHODS Literature reviews of the PubMed, EMBASE and CENTRAL databases were conducted to identify randomized controlled trials (RCTs) comparing endothelin-receptor antagonist treatment with placebo in patients with diabetic nephropathy. Quality assessment was performed by using the Cochrane Handbook's tools for assessing risk of bias; meta-analysis was conducted by RevMan 5.3. RESUTLS Five RCTs (n=2034 patients) were included for analysis. Compared with placebo, endothelin-receptor antagonists showed significant benefits for lowering albuminuria (five trials, n=2034 patients; SMD 0.66 95% confidence interval (CI) 0.56 to 0.76), but there was no significant difference in the risk of death (two trials, n=1674 patients; RR 1.49 95% CI 0.81 to 2.76). In addition, risk of cardiovascular events and other serious adverse events were significantly higher in the endothelin-receptor antagonists group than the placebo group (four trials, n=1956 patients; RR 1.45 95% CI 1.07 to 1.97; five trials, n=2034 patients; RR 1.32 95% CI 1.10 to 1.58). CONCLUSION Endothelin-receptor antagonists can reduce albuminuria in patients with diabetic nephropathy, although use resulted in more serious adverse events compared with placebo. There is a potential need for further RCTs, which has larger sample size and longer duration.
Collapse
Affiliation(s)
- Wenming Yuan
- Renal Division, Department of Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Kidney Epidemiology and Cost Center, University of Michigan, Ann Arbor, MI, USA
| | - Ji Wang
- Division of Evidence-Based Medicine and Clinical Epidemiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jing Li
- Division of Evidence-Based Medicine and Clinical Epidemiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Shenju Gou
- Renal Division, Department of Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Ping Fu
- Renal Division, Department of Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Yasir A, Hardigan T, Ergul A. Diabetes-mediated middle cerebral artery remodeling is restored by linagliptin: Interaction with the vascular smooth muscle cell endothelin system. Life Sci 2016; 159:76-82. [PMID: 26944436 DOI: 10.1016/j.lfs.2016.02.096] [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: 01/09/2016] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Endothelin-1 (ET-1) mediates cerebrovascular remodeling in vascular smooth muscle layer of the middle cerebral arteries (MCA) in type-2 diabetic Goto-Kakizaki (GK) rats. While metformin, oral glucose lowering agent, prevent/restores vascular remodeling and reduce systemic and local ET-1 levels whether this effect was specific to metformin remained unknown. Our working hypotheses were 1) linagliptin, a DPP-IV inhibitor, can reverse diabetes-mediated cerebrovascular remodeling and this is associated with decreased ET-1, and 2) linagliptin prevents the high glucose induced increase in ET-1 and ET receptors in brain vascular smooth muscle cells (bVSMCs). METHODS Diabetic and non-diabetic GK rats were treated with linagliptin (4weeks). MCAs were fixed in buffered 4% paraformaldehyde and used for morphometry. Human bVSMCs incubated in normal glucose (5.5mM)/high glucose (25mM) conditions were treated with the linagliptin (100nM; 24h). ET-1 secretion and ET receptors were measured in media and cell lysate respectively. Immunostaining was performed for ET-A and ET-B receptor. ET receptors were also measured in cells treated with ET-1 (100nM) and linagliptin. RESULTS Linagliptin treatment regressed vascular remodeling of MCAs in diabetic animals but had no effect on blood glucose. bVSMCs in normal/high glucose condition did not show any significant difference in ET-1 secretion or ET-A and ET-B receptor expression. ET-1 treatment in high glucose condition significantly increased the ET-A receptors and this effect was inhibited by linagliptin. CONCLUSIONS Linagliptin is effective in reversing established pathological cerebrovascular remodeling associated with diabetes. Attenuation of the ET system could be a pleiotropic effect of linagliptin that provides vascular protection.
Collapse
Affiliation(s)
- Abdul Yasir
- Charlie Norwood Veterans Administration Medical Center, Augusta University, Augusta, Georgia, United States; Department of Physiology, Augusta University, Augusta, Georgia, United States
| | - Trevor Hardigan
- Department of Physiology, Augusta University, Augusta, Georgia, United States
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta University, Augusta, Georgia, United States; Department of Physiology, Augusta University, Augusta, Georgia, United States.
| |
Collapse
|
27
|
Relationship of endothelin-1 and NLRP3 inflammasome activation in HT22 hippocampal cells in diabetes. Life Sci 2016; 159:97-103. [PMID: 26883974 DOI: 10.1016/j.lfs.2016.02.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 12/22/2022]
Abstract
Diabetes increases the risk and worsens the progression of cognitive decline. Diabetic rats treated with the dual endothelin receptor antagonist bosentan, have been shown to improve hippocampal-based cognitive deficits. The NLRP3 inflammasome has been implicated in vascular complications of diabetes. We hypothesized that diabetes-mediated increase in endothelin-1 (ET-1) in hippocampal cells causes NLRP3 activation and inflammation. An in vitro model was employed by exposing HT22 hippocampal cells to normal (25mM), low (5.5mM) and high (50mM) glucose conditions with and without palmitate (200μM) in the presence and absence of 10μM bosentan for 24h. NLRP3 activity was measured by western blotting for cryopyrin and caspase-1. ET-1 and IL-1β expression was determined by ELISA. HT22 cells synthesize high levels of ET-1 in normal conditions, which was reduced with palmitate and bosentan as well as low and high glucose conditions. Decreased ET-1 levels were associated with greater activation of NLRP3 and IL-1β in normal glucose. High glucose increased NLRP3 markers and activation compared to normal and low glucose. These data suggest that ET-1 may be protective to neurons. Although endothelin antagonism may be beneficial in improving vascular dysfunction and cognitive impairment, its impact on hippocampal neurons should be further explored.
Collapse
|
28
|
Solini A, Rossi C, Duranti E, Taddei S, Natali A, Virdis A. Saxagliptin prevents vascular remodeling and oxidative stress in db/db mice. Role of endothelial nitric oxide synthase uncoupling and cyclooxygenase. Vascul Pharmacol 2016; 76:62-71. [DOI: 10.1016/j.vph.2015.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/23/2015] [Accepted: 10/04/2015] [Indexed: 12/17/2022]
|
29
|
Abdelsaid M, Williams R, Hardigan T, Ergul A. Linagliptin attenuates diabetes-induced cerebral pathological neovascularization in a blood glucose-independent manner: Potential role of ET-1. Life Sci 2015; 159:83-89. [PMID: 26631506 DOI: 10.1016/j.lfs.2015.11.026] [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: 10/05/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 01/01/2023]
Abstract
AIMS We have shown that glycemic control with metformin or endothelin-1 (ET-1) inhibition with bosentan prevents and restores diabetes-mediated cerebral pathological remodeling and neovascularization. Our recent data suggest that linagliptin, a member of the dipeptidyl peptidase-4 inhibitor class of glucose-lowering agents, prevents cerebrovascular remodeling and dysfunction independent of its blood glucose lowering effects. We hypothesized that linagliptin prevents pathological neovascularization via the modulation of the ET-1 system. MATERIALS AND METHODS 24-week old diabetic Goto-Kakizaki and nondiabetic Wistar rats were treated for 4weeks with either vehicle chow or chow containing 166mg/kg linagliptin. At termination, FITC-dextran was injected to visualize the vasculature. Brain sections were imaged by confocal microscopy for vascular density, tortuosity, vascular volume, and surface in both the cortex and striatum. Retinal acellular capillary formation was measured. Brain microvascular endothelial cells (BMVEC) isolated from control or diabetic rats were treated with linagliptin with or without ET-1 dual receptor antagonist and tested for angiogenic properties with cell migration and tube formation assays. KEY FINDING Linagliptin reduced all indices of cerebral neovascularization compared with control rats. In vitro, linagliptin normalized the augmented angiogenic properties of BMVECs isolated from diabetic animals and bosentan reversed this response. Cells from diabetic animals had higher ET-1 and less ETB receptors than in control cells. Linagliptin significantly decreased ET-1 levels and increased ETB receptors. SIGNIFICANCE ET system contributes to pathological neovascularization in diabetes as evidenced by restoration of functional angiogenesis by bosentan treatment and prevention of linagliptin-mediated improvement of angiogenesis in the in vitro model.
Collapse
Affiliation(s)
- Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia.
| | - Raeonda Williams
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Trevor Hardigan
- Department of Physiology, Georgia Regents University, Augusta, Georgia
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia; Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, Georgia; Department of Physiology, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
30
|
Abstract
Systemic sclerosis (SSc) is a multisystem connective tissue disorder featured by vascular injury and fibrosis of the skin and various internal organs with autoimmune background. Although the pathogenesis of SSc still remains elusive, it is generally accepted that initial vascular injury due to autoimmunity and/or environmental factors causes structural and functional abnormalities of vasculature which eventually result in the constitutive activation of fibroblasts in various organs. Structural alterations consist of destructive vasculopathy (loss of small vessels) and proliferative obliterative vasculopathy (occlusion of arterioles and small arteries with fibro-proliferative change) caused by impaired compensatory vasculogenesis and angiogenesis. Impaired function of SSc vasculature includes the altered expression of cell adhesion molecules predominantly inducing Th2 and Th17 cell infiltration, endothelial dysfunction primarily due to the low availability of nitric oxide, the activated endothelial-to-mesenchymal transition leading to fibro-proliferative vascular change and tissue fibrosis, and the impaired coagulation/fibrinolysis system promoting the formation of intravascular fibrin deposits. Recent new insights into the therapeutic mechanisms of intravenous cyclophosphamide pulse and bosentan and the establishment of a new SSc animal model (Klf5 (+/-);Fli1 (+/-) mice) provide us useful clues to further understand the development of vascular alterations characteristic of SSc. This article overviewed the present understanding of the pathogenesis of SSc vasculopathy.
Collapse
|
31
|
Akamata K, Asano Y, Yamashita T, Noda S, Taniguchi T, Takahashi T, Ichimura Y, Toyama T, Trojanowska M, Sato S. Endothelin receptor blockade ameliorates vascular fragility in endothelial cell-specific Fli-1-knockout mice by increasing Fli-1 DNA binding ability. Arthritis Rheumatol 2015; 67:1335-44. [PMID: 25707716 DOI: 10.1002/art.39062] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 02/03/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE It is generally accepted that blockade of endothelin receptors has potentially beneficial effects on vasculopathy associated with systemic sclerosis (SSc). The aim of this study was to clarify the molecular mechanism underlying these effects using endothelial cell-specific Fli-1-knockout (Fli-1 ECKO) mice, an animal model of SSc vasculopathy. METHODS Levels of messenger RNA for target genes and the expression and phosphorylation levels of target proteins were determined in human and murine dermal microvascular endothelial cells by real-time quantitative reverse transcription-polymerase chain reaction and immunoblotting, respectively. The binding of Fli-1 to the target gene promoters was evaluated using chromatin immunoprecipitation. Expression levels of Fli-1 and α-smooth muscle actin in murine skin were evaluated using immunohistochemistry. Vascular structure and permeability were evaluated in mice injected with fluorescein isothiocyanate-dextran and Evans blue dye, respectively. RESULTS In human dermal microvascular endothelial cells, endothelin 1 induced phosphorylation of Fli-1 at Thr(312) through the sequential activation of c-Abl and protein kinase Cδ, leading to a decrease in Fli-1 protein levels as well as a decrease in binding of Fli-1 to the target gene promoters, whereas bosentan treatment reversed those effects. In Fli-1 ECKO mice, 4 weeks of treatment with bosentan increased endothelial Fli-1 expression, resulting in vascular stabilization and the restoration of impaired leaky vessels. CONCLUSION The vascular fragility of Fli-1 ECKO mice was improved by bosentan through the normalization of Fli-1 protein levels and activity in endothelial cells, which may explain, in part, the mechanism underlying the beneficial effects of endothelin receptor blockade on SSc vasculopathy.
Collapse
Affiliation(s)
- Kaname Akamata
- University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abdelsaid M, Prakash R, Li W, Coucha M, Hafez S, Johnson MH, Fagan SC, Ergul A. Metformin treatment in the period after stroke prevents nitrative stress and restores angiogenic signaling in the brain in diabetes. Diabetes 2015; 64:1804-17. [PMID: 25524911 PMCID: PMC4407857 DOI: 10.2337/db14-1423] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/11/2014] [Indexed: 12/04/2022]
Abstract
Diabetes impedes vascular repair and causes vasoregression in the brain after stroke, but mechanisms underlying this response are still unclear. We hypothesized that excess peroxynitrite formation in diabetic ischemia/reperfusion (I/R) injury inactivates the p85 subunit of phosphoinositide 3-kinase (PI3K) by nitration and diverts the PI3K-Akt survival signal to the p38-mitogen-activated protein kinase apoptosis pathway. Nitrotyrosine (NY), Akt and p38 activity, p85 nitration, and caspase-3 cleavage were measured in brains from control, diabetic (GK), or metformin-treated GK rats subjected to sham or stroke surgery and in brain microvascular endothelial cells (BMVECs) from Wistar and GK rats subjected to hypoxia/reoxygenation injury. GK rat brains showed increased NY, caspase-3 cleavage, and p38 activation and decreased Akt activation. Metformin attenuated stroke-induced nitrative signaling in GK rats. GK rat BMVECs showed increased basal nitrative stress compared with controls. A second hit by hypoxia/reoxygenation injury dramatically increased the nitration of p85 and activation of p38 but decreased Akt. These effects were associated with impairment of angiogenic response and were restored by treatment with the peroxynitrite scavenger 5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron III chloride or the nitration inhibitor epicatechin. Our results provide evidence that I/R-induced peroxynitrite inhibits survival, induces apoptosis, and promotes peroxynitrite as a novel therapeutic target for the improvement of reparative angiogenesis after stroke in diabetes.
Collapse
Affiliation(s)
- Mohammed Abdelsaid
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA
| | - Roshini Prakash
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | - Weiguo Li
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA
| | - Maha Coucha
- Department of Physiology, Georgia Regents University, Augusta, GA
| | - Sherif Hafez
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | | | - Susan C Fagan
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| | - Adviye Ergul
- Charlie Norwood Veterans Administration Medical Center, Augusta, GA Department of Physiology, Georgia Regents University, Augusta, GA Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA
| |
Collapse
|
33
|
Singh G, Sharma B, Jaggi AS, Singh N. Efficacy of bosentan, a dual ETA and ETB endothelin receptor antagonist, in experimental diabetes induced vascular endothelial dysfunction and associated dementia in rats. Pharmacol Biochem Behav 2014; 124:27-35. [DOI: 10.1016/j.pbb.2014.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 01/17/2023]
|