1
|
Abu-Alghayth MH, Al-Kuraishy HM, Al-Gareeb AI, Alexiou A, Papadakis M, Bahaa MM, Afifi M, Al-Farga A, Wahsh E, Batiha GES. Atheroprotective role of vinpocetine: an old drug with new indication. Inflammopharmacology 2024; 32:3669-3678. [PMID: 39141151 PMCID: PMC11550280 DOI: 10.1007/s10787-024-01529-5] [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: 11/24/2023] [Accepted: 03/22/2024] [Indexed: 08/15/2024]
Abstract
Endothelial dysfunction is considered one of the main causes of atherosclerosis and elevated blood pressure. Atherosclerosis (AS) formation is enhanced by different mechanisms including cytokine generation, vascular smooth muscle cell proliferation, and migration. One of the recent treatment toward endothelial dysfunction is vinpocetine (VPN). VPN is an ethyl apovincaminate used in the management of different cerebrovascular disorders and endothelial dysfunction through inhibition of atherosclerosis formation. VPN is a potent inhibitor of phosphodiesterase enzyme 1 (PDE1) as well it has anti-inflammatory and antioxidant effects through inhibition of the expression of nuclear factor kappa B (NF-κB). VPN has been shown to be effective against development and progression of AS. However, the underlying molecular mechanism was not fully clarified. Consequently, objective of the present narrative review was to clarify the mechanistic role of VPN in AS. Most of pro-inflammatory cytokines released from macrophages are inhibited by the action of VPN via NF-κB-dependent mechanism. VPN blocks monocyte adhesion and migration by inhibiting the expression of pro-inflammatory cytokines. As well, VPN is effective in reducing oxidative stress, a cornerstone in the pathogenesis of AS, through inhibition of NF-κB and PDE1. VPN promotes plaque stability and prevent erosion and rupture of atherosclerotic plaque. In conclusion, VPN through mitigation of inflammatory and oxidative stress with plaque stability effects could be effective agent in the management of endothelial dysfunction through inhibition of atherosclerosis mediators.
Collapse
Affiliation(s)
- Mohammed H Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, 67714, Bisha, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Athanasios Alexiou
- University Centre for Research and Development, Chandigarh University, Chandigarh-Ludhiana highway, Mohali, Punjab, India
- Department of Research and Development, Funogen, 11741, Athens, Greece
- Department of Research and Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mostafa M Bahaa
- Pharmacy Practice Department, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mohammed Afifi
- Department of Biochemistry, College of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Ammar Al-Farga
- Department of Biochemistry, College of Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Eman Wahsh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University, Arish Campus, Arish, 45511, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
2
|
Bahreiny SS, Ahangarpour A, Aghaei M, Mohammadpour Fard R, Jalali Far MA, Sakhavarz T. A closer look at Galectin-3: its association with gestational diabetes mellitus revealed by systematic review and meta-analysis. J Diabetes Metab Disord 2024; 23:1621-1633. [PMID: 39610475 PMCID: PMC11599495 DOI: 10.1007/s40200-024-01461-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/20/2024] [Indexed: 11/30/2024]
Abstract
Purpose Gestational diabetes mellitus (GDM) represents a significant metabolic disorder that affects pregnant women worldwide and has negative consequences for both the mother and her offspring. This research aims to investigate the relation between circulating levels of Galectin-3 and the incidence of GDM, and to evaluate its potential as a biomarker for monitoring and early detection of the disease. Methods A thorough search of the literature has been performed using databases such as Scopus, Web of science, Embase, Cochrane Library and PubMed. The standardized mean difference (SMD) and corresponding confidence intervals (CIs) were used to compute the effect size from individual records and pooled using the Random-effect model. Results Our meta-analysis synthesized data from 9 studies, encompassing 1,286 participants (533 GDM patients and 753 healthy pregnant controls). The findings demonstrated a considerable increase in Galectin-3 levels among individuals diagnosed with GDM as compared to the healthy control (SMD = 0.929; CI: 0.179-1.679; p = 0.015), with observed heterogeneity (I2 = 87%; p < 0.001). Subgroup analyses revealed the influence of factors such as age, BMI, study design, and sample type on Galectin-3 levels. A meta-regression analysis further identified trends indicating that levels of Galectin-3 are linked to gestational age, specific geographical areas, and sample size. Conclusion Increased levels of Galectin-3 exhibit a significant association with GDM, indicating its prospective utility as a biomarker for early detection and risk assessment. Further research is warranted to elucidate its regulation and clinical implications in GDM management. Graphical Abstract Supplementary Information The online version contains supplementary material available at 10.1007/s40200-024-01461-z.
Collapse
Affiliation(s)
- Seyed Sobhan Bahreiny
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Mohammadpour Fard
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ali Jalali Far
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tannaz Sakhavarz
- Department of Biochemistry, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| |
Collapse
|
3
|
Wan Q, Lu Q, Luo S, Guan C, Zhang H. The beneficial health effects of puerarin in the treatment of cardiovascular diseases: from mechanisms to therapeutics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7273-7296. [PMID: 38709267 DOI: 10.1007/s00210-024-03142-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death globally that seriously threaten human health. Although novel western medicines have continued to be discovered over the past few decades to inhibit the progression of CVDs, new drug research and development for treating CVDs with less side effects and adverse reactions are continuously being desired. Puerarin is a natural product found in a variety of medicinal plants belonging to the flavonoid family with potent biological and pharmacological activities. Abundant research findings in the literature have suggested that puerarin possesses a promising prospect in treating CVDs. In recent years, numerous new molecular mechanisms of puerarin have been explored in experimental and clinical studies, providing new evidence for this plant metabolite to protect against CVDs. This article systematically introduces the history of use, bioavailability, and various dosage forms of puerarin and further summarizes recently published data on the major research advances and their underlying therapeutic mechanisms in treating CVDs. It may provide references for researchers in the fields of pharmacology, natural products, and internal medicine.
Collapse
Affiliation(s)
- Qiang Wan
- Affiliated Hospital of Jiangxi University of Chinese Medicine, 445 Bayi Avenue, Nanchang, 330006, China.
- Clinical Medical College, Jiangxi University of Chinese Medicine, 445 Bayi Avenue, Nanchang, 330006, China.
| | - Qiwen Lu
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Sang Luo
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Chengyan Guan
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| | - Hao Zhang
- Graduate School, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang, 330004, China
| |
Collapse
|
4
|
Jung S, Cheong S, Lee Y, Lee J, Lee J, Kwon MS, Oh YS, Kim T, Ha S, Kim SJ, Jo DH, Ko J, Jeon NL. Integrating Vascular Phenotypic and Proteomic Analysis in an Open Microfluidic Platform. ACS NANO 2024; 18:24909-24928. [PMID: 39208278 PMCID: PMC11394367 DOI: 10.1021/acsnano.4c05537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
This research introduces a vascular phenotypic and proteomic analysis (VPT) platform designed to perform high-throughput experiments on vascular development. The VPT platform utilizes an open-channel configuration that facilitates angiogenesis by precise alignment of endothelial cells, allowing for a 3D morphological examination and protein analysis. We study the effects of antiangiogenic agents─bevacizumab, ramucirumab, cabozantinib, regorafenib, wortmannin, chloroquine, and paclitaxel─on cytoskeletal integrity and angiogenic sprouting, observing an approximately 50% reduction in sprouting at higher drug concentrations. Precise LC-MS/MS analyses reveal global protein expression changes in response to four of these drugs, providing insights into the signaling pathways related to the cell cycle, cytoskeleton, cellular senescence, and angiogenesis. Our findings emphasize the intricate relationship between cytoskeletal alterations and angiogenic responses, underlining the significance of integrating morphological and proteomic data for a comprehensive understanding of angiogenesis. The VPT platform not only advances our understanding of drug impacts on vascular biology but also offers a versatile tool for analyzing proteome and morphological features across various models beyond blood vessels.
Collapse
Affiliation(s)
- Sangmin Jung
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Sunghun Cheong
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Yoonho Lee
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jungseub Lee
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jihye Lee
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Min-Seok Kwon
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
- Department
of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Sun Oh
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Taewan Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungjae Ha
- ProvaLabs,
Inc., Seoul 08826, Republic of Korea
| | - Sung Jae Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT
Foundry, Seoul National University, Seoul 08826, Republic of Korea
- Inter-university
Semiconductor Research Center, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Dong Hyun Jo
- Department
of Anatomy and Cell Biology, Seoul National
University College of Medicine, Seoul 03080, Republic of Korea
| | - Jihoon Ko
- Department
of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic
of Korea
| | - Noo Li Jeon
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Institute
of Advanced Machines and Design, Seoul National
University, Seoul 08826, Republic
of Korea
- Qureator, Inc., San
Diego, California 92121, United States
| |
Collapse
|
5
|
Ashok K, Martinez T, Sesen J, Nasim S, Lang SS, Heuer G, Tucker A, Lopez-Ramirez MA, Smith ER, Ghalali A. Lectin-type oxidized LDL receptor-1 as a potential therapeutic target for cerebral cavernous malformations treatment. Front Neurosci 2024; 18:1442110. [PMID: 39234183 PMCID: PMC11371587 DOI: 10.3389/fnins.2024.1442110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Cerebral cavernous malformations (CCMs) are pathologic lesions comprised of clusters of thin-walled capillaries characterized by abnormal proliferation, angiogenesis, and bleeding secondary to somatic or germline mutations in endothelial cells. CCMs can cause headaches, seizures and/or neurological defects. There is a clinical need to develop better tools to detect CCMs and follow their progression in conjunction with the current use of neuroimaging techniques. Here we present data supporting the utility of LOX-1 (lectin-type oxidized LDL receptor 1), a 50 kDa transmembrane protein implicated in endothelial cell dysfunction and ischemia, as a putative biomarker for CCM. Methods CCM urine samples (n = 23) were collected from pediatric CCM patients. Matched healthy controls (n = 24) were collected from pediatric patients with either Chiari I malformation or fatty filum terminale, and otherwise normal findings. All samples were collected with patient/family consent and institutional review board approval.Samples were analyzed with Olink Proteomic Proximity Extension Assay (PEA). Differences in expression for 2,925 unique proteins were quantified between healthy control urine samples and CCM urine samples. The results were normalized, validated, and analyzed for demographic bias. In addition to urine samples, CCM tissue from patients was harvested and used to create primary cell lines for in vitro analysis of LOX-1 expression, in addition to immunofluorescence of lesional tissue excised at surgery. Results ANOVA analysis of the CCM urine samples showed a statistically significant increase in LOX-1 compared to the control samples, with CCM patients exhibiting a > 5-fold increase in urinary expression. Corroborating these elevated levels of circulating marker, analysis of source tissue from surgically resected CCMs revealed that LOX-1 is increased in both CCM patient cavernoma primary cell lines and operative specimens. Conclusion LOX-1 is involved with pathways implicated in CCM pathogenesis and our data here reveals that LOX-1 expression is significantly elevated in CCM patients as compared to matched healthy control individuals, including both source tissue from surgically excised CCMs and in analysis of samples collected from outside of the central nervous system, particularly urine. This proof-of-principle data suggests that LOX-1 may have potential utility as a target for CCM treatment and supports further investigation related to its potential mechanistic impact on CCM pathogenesis.
Collapse
Affiliation(s)
- Karthik Ashok
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Tyra Martinez
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Julie Sesen
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Sana Nasim
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
| | - Shih-Shan Lang
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Gregory Heuer
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander Tucker
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Edward R Smith
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, United States
| | - Aram Ghalali
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Al-Kuraishy HM, Al-Gareeb AI, Elekhnawy E, Batiha GES. Possible role of LCZ696 in atherosclerosis: new inroads and perspective. Mol Cell Biochem 2024; 479:1895-1908. [PMID: 37526794 DOI: 10.1007/s11010-023-04816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/15/2023] [Indexed: 08/02/2023]
Abstract
LCZ696 blocks both angiotensin receptor type 1 (ATR1) and neprilysin (NEP), which are intricate in the degradation of natriuretic peptides (NPs) and other endogenous peptides. It has been shown NEP inhibitors and LCZ696 could be effectively in the management of atherosclerosis (AS). However, the underlying mechanism of LCZ696 in AS is needed to be clarified entirely. Hence, this review is directed to reconnoiter the mechanistic role of LCZ696 in AS. The anti-inflammatory role of LCZ696 is related to the inhibition of transforming growth factor beta (TGF-β)-activated kinase 1 (TAK) and nod-like receptor pyrin 3 receptor (NLRP3) inflammasome. Moreover, LCZ696, via inhibition of pro-inflammatory cytokines, oxidative stress, apoptosis and endothelial dysfunction can attenuate the development and progression of AS. In conclusion, LCZ696 could be effective in the management of AS through modulation of inflammatory and oxidative signaling. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, AL-Mustansiriyia University, Baghdad, Iraq
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AL Beheira, Egypt.
| |
Collapse
|
7
|
Ma Y, Han J, Wang K, Han H, Hu Y, Li H, Wu S, Zhang L. Research progress of Ganoderma lucidum polysaccharide in prevention and treatment of Atherosclerosis. Heliyon 2024; 10:e33307. [PMID: 39022015 PMCID: PMC11253544 DOI: 10.1016/j.heliyon.2024.e33307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease resulting from dysregulated lipid metabolism, constituting the pathophysiological foundation of cardiovascular and cerebrovascular diseases. AS has a high incidence rate and mortality rate worldwide. As such, traditional Chinese medicine (TCM) has been widely used recently due to its stable therapeutic effect and high safety. Ganoderma lucidum polysaccharides (GLP) are the main active ingredients of Ganoderma lucidum, a Chinese herbal medicine. Research has also shown that GLP has anti-inflammatory and antioxidant properties, regulates gut microbiota, improves blood glucose and lipid levels, and inhibits obesity. Most of the current research on GLP anti-AS is focused on animal models. Thus, its clinical application remains to be discovered. In this review, we combine relevant research results and start with the pathogenesis and risk factors of GLP on AS, proving that GLP can prevent and treat AS, providing a scientific basis and reference for the future prevention and treatment of AS with GLP.
Collapse
Affiliation(s)
- YiZheng Ma
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, Jinan, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| | - JingBo Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - KangFeng Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| | - Huan Han
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, Jinan, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| | - YiBin Hu
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, Jinan, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| | - He Li
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, 250355, Jinan, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| | - ShengXian Wu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - LiJuan Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, 250014, Jinan, China
| |
Collapse
|
8
|
Kuehu DL, Fu Y, Nasu M, Yang H, Khadka VS, Deng Y. Use of Microalgae-Derived Astaxanthin to Improve Cytoprotective Capacity in the Ileum of Heat-Induced Oxidative Stressed Broilers. Animals (Basel) 2024; 14:1932. [PMID: 38998041 PMCID: PMC11240551 DOI: 10.3390/ani14131932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
The gastrointestinal tract has a pivotal role in nutrient absorption, immune function, and overall homeostasis. The ileum segment of the small intestine plays respective roles in nutrient breakdown and absorption. The purpose of this study was to investigate the impact of heat-induced oxidative stress and the potential mitigating effects of an astaxanthin antioxidant treatment on the ileum of broilers. By comparing the growth performance and gene expression profiles among three groups-thermal neutral, heat stress, and heat stress with astaxanthin-thermal neutral temperature conditions of 21-22 °C and heat stress temperature of 32-35 °C, this research aims to elucidate the role of astaxanthin in supporting homeostasis and cellular protection in the ileum. Results showed both treatments under heat stress experienced reduced growth performance, while the group treated with astaxanthin showed a slightly lesser decline. Results further showed the astaxanthin treatment group significantly upregulated in the cytoprotective gene expression for HSF2, SOD2, GPX3, and TXN, as well as the upregulation of epithelial integrity genes LOX, CLDN1, and MUC2. In conclusion, our experimental findings demonstrate upregulation of cytoprotective and epithelial integrity genes, suggesting astaxanthin may effectively enhance the cellular response to heat stress to mitigate oxidative damage and contribute to cytoprotective capacity.
Collapse
Affiliation(s)
- Donna Lee Kuehu
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
- Department of Molecular Biosciences and Bioengineering, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI 96822, USA
| | - Yuanyuan Fu
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
| | - Masaki Nasu
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
| | - Hua Yang
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
| | - Vedbar S. Khadka
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
| | - Youping Deng
- Bioinformatics Core, Department of Quantitative Health Sciences, John A Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA; (D.L.K.); (Y.F.); (M.N.); (H.Y.); (V.S.K.)
| |
Collapse
|
9
|
Alshehri AA, Al-Kuraishy HM, Al-Gareeb AI, Jawad SF, Khawagi WY, Alexiou A, Papadakis M, Assiri AA, Elhadad H, El-Saber Batiha G. The anti-inflammatory properties of vinpocetine mediates its therapeutic potential in management of atherosclerosis. J Inflamm (Lond) 2024; 21:19. [PMID: 38858751 PMCID: PMC11165849 DOI: 10.1186/s12950-024-00394-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
Atherosclerosis (AS) formation is enhanced by different mechanisms including cytokine generation, vascular smooth muscle cell proliferation, and migration. One of the recent treatments towards endothelial dysfunction and AS is Vinpocetine (VPN). VPN is a potent inhibitor of phosphodiesterase enzyme 1 (PDE-1) and has anti-inflammatory and antioxidant effects through inhibition the expression of nuclear factor kappa B (NF-κB). VPN has been shown to be effective against the development and progression of AS. However, the underlying molecular mechanism was not fully clarified. Consequently, objective of the present review was to discuss the mechanistic role of VPN in the pathogenesis AS. Most of pro-inflammatory cytokines that released from macrophages are inhibited by action of VPN through NF-κB-dependent mechanism. VPN blocks monocyte adhesion and migration by constraining the expression and action of pro-inflammatory cytokines. As well, VPN is effective in reducing of oxidative stress a cornerstone in the pathogenesis of AS through inhibition of NF-κB and PDE1. VPN promotes plaque stability and prevents the erosion and rupture of atherosclerotic plaque. In conclusion, VPN through mitigation of inflammatory and oxidative stress, and improvement of plaque stability effects could be effective agent in the management of AS.
Collapse
Affiliation(s)
- Abdullah A Alshehri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Al Huwaya, Taif, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir ibn Hayyan Medical University, PO.Box13, Al-Ameer Qu./Najaf, Iraq
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, 51001, Iraq
| | - Wael Y Khawagi
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Al Huwaya, Taif, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, Wien, 1030, Austria
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, 11741, Greece
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Universityof Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Abdullah A Assiri
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University Abha, Abha, Saudi Arabia
| | - Heba Elhadad
- Department of Parasitology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
10
|
Yu SS, Du JL. Current views on selenoprotein S in the pathophysiological processes of diabetes-induced atherosclerosis: potential therapeutics and underlying biomarkers. Diabetol Metab Syndr 2024; 16:5. [PMID: 38172976 PMCID: PMC10763436 DOI: 10.1186/s13098-023-01247-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) consistently ranks as the primary mortality factor among diabetic people. A thorough comprehension of the pathophysiological routes and processes activated by atherosclerosis (AS) caused by diabetes mellitus (DM), together with the recognition of new contributing factors, could lead to the discovery of crucial biomarkers and the development of innovative drugs against atherosclerosis. Selenoprotein S (SELENOS) has been implicated in the pathology and progression of numerous conditions, including diabetes, dyslipidemia, obesity, and insulin resistance (IR)-all recognized contributors to endothelial dysfunction (ED), a precursor event to diabetes-induced AS. Hepatic-specific deletion of SELENOS accelerated the onset and progression of obesity, impaired glucose tolerance and insulin sensitivity, and increased hepatic triglycerides (TG) and diacylglycerol (DAG) accumulation; SELENOS expression in subcutaneous and omental adipose tissue was elevated in obese human subjects, and act as a positive regulator for adipogenesis in 3T3-L1 preadipocytes; knockdown of SELENOS in Min6 β-cells induced β-cell apoptosis and reduced cell proliferation. SELENOS also participates in the early stages of AS, notably by enhancing endothelial function, curbing the expression of adhesion molecules, and lessening leukocyte recruitment-actions that collectively reduce the formation of foam cells. Furthermore, SELENOS forestalls the apoptosis of vascular smooth muscle cells (VSMCs) and macrophages, mitigates vascular calcification, and alleviates inflammation in macrophages and CD4+ T cells. These actions help stifle the creation of unstable plaque characterized by thinner fibrous caps, larger necrotic cores, heightened inflammation, and more extensive vascular calcification-features seen in advanced atherosclerotic lesion development. Additionally, serum SELENOS could function as a potential biomarker, and SELENOS single nucleotide polymorphisms (SNPs) rs4965814, rs28628459, and rs9806366, might be effective gene markers for atherosclerosis-related diseases in diabetes. This review accentuates the pathophysiological processes of atherosclerosis in diabetes and amasses current evidence on SELENOS's potential therapeutic benefits or as predictive biomarkers in the various stages of diabetes-induced atherosclerosis.
Collapse
Affiliation(s)
- Shan-Shan Yu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China
| | - Jian-Ling Du
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China.
| |
Collapse
|
11
|
Zhou Z, Feng Z, Sun X, Wang Y, Dou G. The Role of Galectin-3 in Retinal Degeneration and Other Ocular Diseases: A Potential Novel Biomarker and Therapeutic Target. Int J Mol Sci 2023; 24:15516. [PMID: 37958500 PMCID: PMC10649114 DOI: 10.3390/ijms242115516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Galectin-3 is the most studied member of the Galectin family, with a large range of mediation in biological activities such as cell growth, proliferation, apoptosis, differentiation, cell adhesion, and tissue repair, as well as in pathological processes such as inflammation, tissue fibrosis, and angiogenesis. As is known to all, inflammation, aberrant cell apoptosis, and neovascularization are the main pathophysiological processes in retinal degeneration and many ocular diseases. Therefore, the review aims to conclude the role of Gal3 in the retinal degeneration of various diseases as well as the occurrence and development of the diseases and discuss its molecular mechanisms according to research in systemic diseases. At the same time, we summarized the predictive role of Gal3 as a biomarker and the clinical application of its inhibitors to discuss the possibility of Gal3 as a novel target for the treatment of ocular diseases.
Collapse
Affiliation(s)
| | | | | | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| | - Guorui Dou
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, China; (Z.Z.); (Z.F.); (X.S.)
| |
Collapse
|
12
|
Ewees MGED, Orfali R, Rateb EE, Hassan HM, Hozzein WN, Alkhalfah DHM, Sree HTA, Abdel Rahman FEZS, Rateb ME, Mahmoud NI. Modulation of mi-RNA25/Ox-LDL/NOX4 signaling pathway by polyphenolic compound Hydroxytyrosol as a new avenue to alleviate cisplatin-induced acute kidney injury, a mechanistic study in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 103:104262. [PMID: 37699441 DOI: 10.1016/j.etap.2023.104262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/16/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023]
Abstract
Acute kidney injury (AKI) caused by Cis is considered one of the most severe adverse effects, which restricts its use and efficacy. This study seeks to examine the potential reno-protective impact of phenolic compound Hydroxytyrosol (HT) against Cis-induced AKI and the possible involvement of the mi-RNA25/Ox-LDL/NOX4 pathway elucidating the probable implicated molecular mechanisms. Forty rats were placed into 5 groups. Group I received saline only. Group II received Cis only. Group III, IV, and V received 20, 50, and 100 mg/kg b.w, of HT, respectively, with Cis delivery. NOX4, Ox-LDL, and gene expression of mi-RNA 25, TNF-α, and HO-1 in renal tissue were detected. HT showed reno-protective effect and significantly upregulated mi-RNA 25 and HO-1 as well as decreased the expression of NOX4, Ox-LDL, and TNF-α. In conclusion, HT may be promising in the fight against Cis-induced AKI through modulation of mi-RNA25/Ox-LDL/NOX4 pathway.
Collapse
Affiliation(s)
- Mohamed Gamal El-Din Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, PO Box 2457, Riyadh 11451, Saudi Arabia.
| | - Enas Ezzat Rateb
- Department of Physiology, Faculty of Medicine, Beni-Suef University, Beni-Suef 62521, Egypt.
| | - Hossam M Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| | - Wael N Hozzein
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| | - Dalal Hussien M Alkhalfah
- Department of Biology. College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia.
| | - Haidy Tamer Abo Sree
- Department of Basic Sciences Department, Biochemistry, Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef 11787, Egypt.
| | - Fatema El-Zahraa S Abdel Rahman
- Department of Basic Sciences Department, Physiology, Faculty of Oral and Dental Medicine, Nahda University, Beni-Suef 11787, Egypt.
| | - Mostafa E Rateb
- School of Computing, Engineering & Physical Sciences, University of the West of Scotland, Paisley PA1 2BE, UK.
| | - Nesreen Ishak Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt.
| |
Collapse
|
13
|
Mayyas F, Ibrahim K. Evaluating Plasma Galectin-3 Levels in Patients With an Increased Risk of Atherosclerotic Cardiovascular Disease Who Underwent Coronary Artery Revascularization. Am J Cardiol 2023; 203:73-80. [PMID: 37481815 DOI: 10.1016/j.amjcard.2023.06.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023]
Abstract
Coronary artery disease (CAD) is a common atherosclerotic cardiovascular disease (ASCVD) associated with significant mortality. Galectin-3 is a novel inflammatory factor implicated in the initiation and progression of atherosclerosis. We aimed to evaluate the association of plasma galectin-3 with the risk of ASCVD and the need for coronary artery revascularization. Patients with angina who underwent coronary angiography were divided into groups per their risk of ASCVD. Patients (n = 385) were stratified into having low (n = 21), moderate (n = 40), high (n = 41), and very high risk (n = 283) for ASCVD. The mean age ± standard error of the mean was 53.9 ± 0.5 years and 73% of patients were men. Plasma galectin-3 levels were higher in patients with CAD than non-CAD primarily in patients with stable and unstable angina. Patients with stable CAD had higher levels of galectin-3 relative to acute coronary syndrome patients. Increased plasma galectin-3 level was associated with increased risk of ASCVD and degree of coronary stenosis. By multivariate analysis, the plasma galectin-3 level was independently associated with increased ASCVD risk and body mass index. Plasma galectin-3 levels were independently higher in patients who underwent percutaneous coronary intervention (PCI) than medically treated patients. In addition, age, male gender, smoking, and diabetes mellitus were associated with PCI. In conclusion, plasma galectin-3 levels are elevated in patients with CAD and associated with increased risk of ASCVD and the need for PCI. Plasma galectin-3 could be used as a potential improving predictor of ASCVD risk and when making therapeutic guidance or selecting patients who underwent PCI when the decision is difficult.
Collapse
Affiliation(s)
- Fadia Mayyas
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan.
| | - Khalid Ibrahim
- Division of Cardiac Surgery, Department of General Surgery, Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
14
|
Xue J, Zhang Z, Sun Y, Jin D, Guo L, Li X, Zhao D, Feng X, Qi W, Zhu H. Research Progress and Molecular Mechanisms of Endothelial Cells Inflammation in Vascular-Related Diseases. J Inflamm Res 2023; 16:3593-3617. [PMID: 37641702 PMCID: PMC10460614 DOI: 10.2147/jir.s418166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Endothelial cells (ECs) are widely distributed inside the vascular network, forming a vital barrier between the bloodstream and the walls of blood vessels. These versatile cells serve myriad functions, including the regulation of vascular tension and the management of hemostasis and thrombosis. Inflammation constitutes a cascade of biological responses incited by biological, chemical, or physical stimuli. While inflammation is inherently a protective mechanism, dysregulated inflammation can precipitate a host of vascular pathologies. ECs play a critical role in the genesis and progression of vascular inflammation, which has been implicated in the etiology of numerous vascular disorders, such as atherosclerosis, cardiovascular diseases, respiratory diseases, diabetes mellitus, and sepsis. Upon activation, ECs secrete potent inflammatory mediators that elicit both innate and adaptive immune reactions, culminating in inflammation. To date, no comprehensive and nuanced account of the research progress concerning ECs and inflammation in vascular-related maladies exists. Consequently, this review endeavors to synthesize the contributions of ECs to inflammatory processes, delineate the molecular signaling pathways involved in regulation, and categorize and consolidate the various models and treatment strategies for vascular-related diseases. It is our aspiration that this review furnishes cogent experimental evidence supporting the established link between endothelial inflammation and vascular-related pathologies, offers a theoretical foundation for clinical investigations, and imparts valuable insights for the development of therapeutic agents targeting these diseases.
Collapse
Affiliation(s)
- Jiaojiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yuting Sun
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Di Jin
- Department of Nephrology, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Liming Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Xiaochun Feng
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Biomacromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Haoyu Zhu
- Department of Nephropathy and Rheumatology in Children, Children’s Medical Center, First Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| |
Collapse
|
15
|
Huang PY, Huang CS, Lin YL, Chen YH, Hung SC, Tsai JP, Hsu BG. Positive Association of Serum Galectin-3 with the Development of Aortic Stiffness of Patients on Peritoneal Dialysis. J Clin Med 2023; 12:jcm12103519. [PMID: 37240626 DOI: 10.3390/jcm12103519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/09/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
A novel cardiovascular stress biomarker known as galectin-3 might be useful for anticipating adverse cardiovascular outcomes. The objective of the current investigation was to assess the association between serum galectin-3 levels and aortic stiffness (AS) in 196 patients on peritoneal dialysis. An enzyme-linked immunosorbent examination and a cuff-based volumetric displacement were employed to determine the levels of serum galectin-3 and the carotid-femoral pulse wave velocity (cfPWV), respectively. The AS group had 48 patients in total (24.5%) with cfPWV greater than 10 m/s. The AS group, when compared with the group without AS, had a significantly higher prevalence of diabetes mellitus and hypertension in addition to greater fasting glucose levels, waist circumference, systolic blood pressure, and serum galectin-3 levels. Multivariate logistic and linear regression analysis demonstrated that serum glactin-3 levels, in addition to gender and age, were significantly and independently associated with cfPWV and AS. Serum galectin-3 levels were linked with AS, according to a receiver operating characteristic curve analysis, with an area under the curve of 0.648 (95% confidence interval, 0.576-0.714; p = 0.0018). In summary, there was a significant correlation between serum galectin-3 levels and cfPWV in patients undergoing peritoneal dialysis therapy for end-stage kidney disease.
Collapse
Affiliation(s)
- Po-Yu Huang
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Chen-Sen Huang
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
| | - Yu-Li Lin
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Yi-Hsin Chen
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 40201, Taiwan
| | - Szu-Chun Hung
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei 23142, Taiwan
| | - Jen-Pi Tsai
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Bang-Gee Hsu
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| |
Collapse
|
16
|
Wang Z, Gao Z, Zheng Y, Kou J, Song D, Yu X, Dong B, Chen T, Yang Y, Gao X, Wang Q, Ye T, Yang W, Zhang X, Li H, Yang L. Melatonin inhibits atherosclerosis progression via galectin-3 downregulation to enhance autophagy and inhibit inflammation. J Pineal Res 2023; 74:e12855. [PMID: 36692032 DOI: 10.1111/jpi.12855] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/24/2022] [Accepted: 01/19/2023] [Indexed: 01/25/2023]
Abstract
Autophagy deficiency in macrophages exacerbates inflammation in atherosclerosis (AS), and recently, galectin-3 (Gal-3) has been implicated as a critical promoter of inflammation in AS. Further, melatonin (Mel) exerts an autophagy-promoting effect in many chronic inflammatory diseases. In this study, we aimed to investigate whether Mel inhibits AS progression by downregulating Gal-3 to enhance autophagy and inhibit inflammation. Thus, we performed in vivo and in vitro experiments using high-fat diet (HFD)-fed ApoE-/- mice and THP-1 macrophages, respectively. Smart-seq of AS plaque macrophages revealed that the differentially expressed genes (DEGs) downregulated by Mel were enriched in immune-related processes, and changes in inflammation status were confirmed based on lower levels of proinflammatory factors in Mel-treated HFD-fed ApoE-/- mice and THP-1 macrophages. Further, via transcriptome-based multiscale network pharmacology platform (TMNP), the upstream target genes of the smart-seq DEGs were identified, and Gal-3 showed a high score. Gal-3 was downregulated both in vivo and in vitro by Mel treatment. Besides, the enrichment of the target genes predicted via the TMNP method indicated that autophagy considerably affected the DEGs. Mel treatment as well as Gal-3 knockdown downregulated most inflammatory response-related proteins could attribute to enhancing autophagy. Mechanistically, Mel treatment inhibited Gal-3 leading to lowering the activity of the nuclear transcription factor-kappa B (NF-κB) pathway, and promoting the nuclear localization of transcription factor EB (TFEB). However, increased secretion of Gal-3 activated the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway and impaired autophagy via binding to CD98. Thus, Mel promoted autophagy and restrained inflammation by downregulating Gal-3, implying that it holds promise as a treatment for AS.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ziyu Gao
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- Science and Research Department, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinghong Zheng
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jiayuan Kou
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China
| | - Dan Song
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xue Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Bowen Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Tianzuo Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yan Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xi Gao
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Qianxue Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ting Ye
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Wei Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- School of Public Health, Qiqihar Medical University, Qiqihar, China
| | - Xu Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hong Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University (Daqing), Daqing, China
| |
Collapse
|
17
|
Jiang B, Zhou X, Yang T, Wang L, Feng L, Wang Z, Xu J, Jing W, Wang T, Su H, Yang G, Zhang Z. The role of autophagy in cardiovascular disease: Cross-interference of signaling pathways and underlying therapeutic targets. Front Cardiovasc Med 2023; 10:1088575. [PMID: 37063954 PMCID: PMC10090687 DOI: 10.3389/fcvm.2023.1088575] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Autophagy is a conserved lysosomal pathway for the degradation of cytoplasmic proteins and organelles, which realizes the metabolic needs of cells and the renewal of organelles. Autophagy-related genes (ATGs) are the main molecular mechanisms controlling autophagy, and their functions can coordinate the whole autophagic process. Autophagy can also play a role in cardiovascular disease through several key signaling pathways, including PI3K/Akt/mTOR, IGF/EGF, AMPK/mTOR, MAPKs, p53, Nrf2/p62, Wnt/β-catenin and NF-κB pathways. In this paper, we reviewed the signaling pathway of cross-interference between autophagy and cardiovascular diseases, and analyzed the development status of novel cardiovascular disease treatment by targeting the core molecular mechanism of autophagy as well as the critical signaling pathway. Induction or inhibition of autophagy through molecular mechanisms and signaling pathways can provide therapeutic benefits for patients. Meanwhile, we hope to provide a unique insight into cardiovascular treatment strategies by understanding the molecular mechanism and signaling pathway of crosstalk between autophagy and cardiovascular diseases.
Collapse
Affiliation(s)
- Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xuan Zhou
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Yang
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linlin Wang
- Department of First Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Longfei Feng
- Department of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zheng Wang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jin Xu
- Department of First Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Weiyao Jing
- Department of Acupuncture-Moxibustion and Tuina, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tao Wang
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Haixiang Su
- Research Center for Translational Medicine, Gansu Province Academic Institute for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - GuoWei Yang
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| | - Zheng Zhang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Center for Heart, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Chen D, Du Y, Ye S, Yu J. Astragaloside IV protects against oxidized low-density lipoprotein-induced injury in human umbilical vein endothelial cells via the histone deacetylase 9 (HDAC9)/NF-κB axis. ENVIRONMENTAL TOXICOLOGY 2023; 38:534-544. [PMID: 36322813 DOI: 10.1002/tox.23696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Atherosclerosis is a main cause of multiple cardiovascular diseases, and cell damage of human umbilical vein endothelial cells (HUVECs) was reported to participate in the development of atherosclerosis. In this study, we aimed to study the action of Astragaloside IV (ASV) on AS development using in vitro AS cell model. METHODS MTT assay, EdU staining assay, and flow cytometry were utilized for detection of cell proliferation and apoptosis, respectively. The protein expression of histone deacetylase 9 (HDAC9), Bax, Bcl-2, p-P65, P65, p-IκBα, and IκBα was gaged using western blot. The angiogenesis was evaluated by tube formation assay. The inflammatory response was evaluated by ELISA kits. SOD activity and MDA level were detected using the matched commercial kits. RT-qPCR was used for HDAC9 mRNA expression measurement. RESULTS Oxidized low-density lipoprotein (ox-LDL) significantly repressed cell proliferation, angiogenesis, and enhanced apoptosis, inflammation, and oxidative stress in HUVECs. ASV addition could alleviate ox-LDL-caused cell damage in HUVECs. Moreover, HDAC9 was overexpressed in AS patients and AS cell model. Functionally, HDAC9 knockdown also exhibited the protective role in ox-LDL-treated HUVECs. In addition, ASV treatment protected against ox-LDL-induced damage in HUVECs via targeting HDAC9. ASV could inactivate the NF-κB pathway via regulating HDAC9 in AS cell model. CONCLUSION ASV exerted the protective effects on ox-LDL-induced damage in HUVECs through the HDAC9/NF-κB axis.
Collapse
Affiliation(s)
- Decai Chen
- Department of Vascular Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang, China
| | - Yan Du
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| | - Shouwan Ye
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| | - Jinsong Yu
- Department of Thyroid and Breast Surgery, Nanyang First People's Hospital Affiliated to Henan University, Nanyang Key Laboratory of Thyroid Tumor Prevention and Treatment, Nanyang, China
| |
Collapse
|
19
|
Andrade FEC, Correia-Silva RD, Covre JL, Lice I, Gomes JÁP, Gil CD. Effects of galectin-3 protein on UVA-induced damage in retinal pigment epithelial cells. PHOTOCHEMICAL & PHOTOBIOLOGICAL SCIENCES : OFFICIAL JOURNAL OF THE EUROPEAN PHOTOCHEMISTRY ASSOCIATION AND THE EUROPEAN SOCIETY FOR PHOTOBIOLOGY 2023; 22:21-32. [PMID: 36036336 DOI: 10.1007/s43630-022-00294-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/17/2022] [Indexed: 01/12/2023]
Abstract
Several inflammatory molecules have been suggested as biomarkers of age-related macular degeneration (AMD). Galectin-3 (Gal-3), which has been shown to have a protective role in corneal injury by promoting epithelial cells adhesion and migration to the extracellular matrix, is also highly expressed in the retinal pigment epithelium (RPE) of patients with AMD. This study evaluated the role of Gal-3 in an in vitro model of UVA-induced RPE damage, as a proof-of-concept. ARPE-19 cells (human RPE cell line), were incubated with Gal-3 at 0.5-2.5 µg/mL concentrations prior to UVA irradiation for 15, 30, and 45 min, which resulted in accumulated doses of 2.5, 5, and 7.5 J/cm2, respectively. After 24 h incubation, MTT and LDH assays, immunofluorescence, and ELISA were performed. UVA irradiation for 15, 30, and 45 min proved to reduce viability in 83%, 46%, and 11%, respectively. Based on the latter results, we chose the intermediate dose (5-J/cm2) for further analysis. Pretreatment with Gal-3 at concentrations > 1.5 µg/mL showed to increase the viability of UVA-irradiated cells (~ 75%) compared to untreated cells (64%). Increased levels of cleaved caspase 3, a marker of cell death, were detected in the ARPE cells after UVA irradiation with or without addition of exogenous Gal-3. The inhibitory effect of Gal-3 on UVA-induced cell damage was characterized by decreased ROS levels and increased p38 activation, as detected by fluorescence analysis. In conclusion, our study suggests a photoprotective effect of Gal-3 on RPE by reducing oxidative stress and increasing p38 activation.
Collapse
Affiliation(s)
- Frans E C Andrade
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu 740, Ed. Lemos Torres-3º andar, São Paulo, SP, 04023-900, Brazil
| | - Rebeca D Correia-Silva
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu 740, Ed. Lemos Torres-3º andar, São Paulo, SP, 04023-900, Brazil
| | - Joyce L Covre
- Department of Ophthalmology, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP, 04023-062, Brazil
| | - Izabella Lice
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu 740, Ed. Lemos Torres-3º andar, São Paulo, SP, 04023-900, Brazil
| | - José Álvaro P Gomes
- Department of Ophthalmology, Universidade Federal de São Paulo (UNIFESP), Sao Paulo, SP, 04023-062, Brazil
| | - Cristiane D Gil
- Department of Morphology and Genetics, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu 740, Ed. Lemos Torres-3º andar, São Paulo, SP, 04023-900, Brazil.
| |
Collapse
|
20
|
Mansour AA, Krautter F, Zhi Z, Iqbal AJ, Recio C. The interplay of galectins-1, -3, and -9 in the immune-inflammatory response underlying cardiovascular and metabolic disease. Cardiovasc Diabetol 2022; 21:253. [PMID: 36403025 PMCID: PMC9675972 DOI: 10.1186/s12933-022-01690-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/08/2022] [Indexed: 11/21/2022] Open
Abstract
Galectins are β-galactoside-binding proteins that bind and crosslink molecules via their sugar moieties, forming signaling and adhesion networks involved in cellular communication, differentiation, migration, and survival. Galectins are expressed ubiquitously across immune cells, and their function varies with their tissue-specific and subcellular location. Particularly galectin-1, -3, and -9 are highly expressed by inflammatory cells and are involved in the modulation of several innate and adaptive immune responses. Modulation in the expression of these proteins accompany major processes in cardiovascular diseases and metabolic disorders, such as atherosclerosis, thrombosis, obesity, and diabetes, making them attractive therapeutic targets. In this review we consider the broad cellular activities ascribed to galectin-1, -3, and -9, highlighting those linked to the progression of different inflammatory driven pathologies in the context of cardiovascular and metabolic disease, to better understand their mechanism of action and provide new insights into the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Franziska Krautter
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zhaogong Zhi
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | - Carlota Recio
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Farmacología Molecular y Traslacional -BIOPharm, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Las Palmas, Spain.
| |
Collapse
|
21
|
Ben Ammar R, Mohamed ME, Alfwuaires M, Abdulaziz Alamer S, Bani Ismail M, Veeraraghavan VP, Sekar AK, Ksouri R, Rajendran P. Anti-Inflammatory Activity of Geraniol Isolated from Lemon Grass on Ox-LDL-Stimulated Endothelial Cells by Upregulation of Heme Oxygenase-1 via PI3K/Akt and Nrf-2 Signaling Pathways. Nutrients 2022; 14:4817. [PMID: 36432506 PMCID: PMC9695721 DOI: 10.3390/nu14224817] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/06/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Among the world's leading causes of cardiovascular disease, atherosclerosis is a chronic inflammatory disorder that affects the arteries. Both vasodilation and vasoconstriction, low levels of nitric oxide and high levels of reactive oxygen species and pro-inflammatory factors characterize dysfunctional blood vessels. Hypertension, and atherosclerosis, all start with this dysfunction. Geraniol, a compound of acyclic monoterpene alcohol, found in plants such as geranium, lemongrass and rose, is a primary constituent of essential oils. It shows a variety of pharmacological properties. This study aimed to investigate the impact of geraniol on Ox-LDL-induced stress and inflammation in human umbilical vein endothelial cells. In this study, HUVECs were treated with Ox-LDL or geraniol at different dose concentrations. MTT assay, Western blot, ROS generation and DNA fragmentation were used to evaluate geraniol's effects on Ox-LDL-induced HUVECs inflammation. The results show that geraniol pre-incubation ameliorates Ox-LDL-mediated HUVECs cytotoxicity and DNA fragmentation. The geraniol inhibited the production of pro-inflammatory cytokines by Ox-LDL, including TNF-α, IL-6 and IL-1β. In Ox-LDL-stimulated HUVECs, geraniol suppresses the nuclear translocation and activity of NF-ᴋB as well as phosphorylation of IkBα. Moreover, geraniol activated the PI3K/AKT/NRF2 pathway in HUVECs, resulting in an increase in the expression of HO-1. Taking our data together, we can conclude that, in HUVECs, geraniol inhibits Ox-LDL-induced inflammation and oxidative stress by targeting PI3/AKT/NRF2.
Collapse
Affiliation(s)
- Rebai Ben Ammar
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria, P.O. Box 901, Hammam-Lif 2050, Tunisia
| | - Maged Elsayed Mohamed
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, University of Zagazig, Zagazig 44519, Egypt
| | - Manal Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Sarah Abdulaziz Alamer
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
| | - Mohammad Bani Ismail
- Department of Basic Medical Sciences, School of Medicine, Aqaba Medical Sciences University, Aqaba 11191, Jordan
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Ashok Kumar Sekar
- Centre for Biotechnology, Anna University, Chennai 600025, Tamil Nadu, India
| | - Riadh Ksouri
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria, P.O. Box 901, Hammam-Lif 2050, Tunisia
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| |
Collapse
|
22
|
Zhang S, Xie S, Gao Y, Wang Y. Triptolide alleviates ox-LDL-induced endothelial inflammation via attenuating the oxidative stress-mediated NF-κB pathway. Curr Ther Res Clin Exp 2022; 97:100683. [PMID: 35989981 PMCID: PMC9385561 DOI: 10.1016/j.curtheres.2022.100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022] Open
Abstract
Background Endothelial inflammation triggered by oxidized LDL (ox-LDL) is a crucial mechanism involved in atherosclerosis. Triptolide (TP), a primary active ingredient of the traditional Chinese medicine Tripterygium wilfordii Hook F, possesses antioxidant and anti-inflammatory properties in vivo. However, limited information is available regarding these effects on endothelial inflammation occurring in atherosclerosis. Objectives This study investigated the effects and possible mechanisms of action of TP on ox–LDL-induced inflammatory responses in human umbilical vein endothelial cells. Methods Human umbilical vein endothelial cells were preincubated with TP at the indicated concentrations for 1 hour and then incubated with ox-LDL (50 µg/mL) for the indicated times. Results Preincubation of cultured human umbilical vein endothelial cells with TP inhibited ox–LDL-induced cytokine and chemokine production, adhesion molecule expression, and monocyte adhesion in a concentration-dependent manner. The concentrations of 8-isoprostane, malondialdehyde, and superoxide increased after human umbilical vein endothelial cells were exposed to ox-LDL, which were associated with decreased activities of total superoxide dismutase and its isoenzyme (ie, CuZn- superoxide dismutase). Preincubation with TP reversed ox–LDL-induced effects in all events. Moreover, preincubation with TP also attenuated ox–LDL-induced nuclear factor-kappa B transcriptional activation in a concentration-dependent manner, via the suppression of inhibitor of kappa Balpha (IκBα) phosphorylation and subsequent nuclear factor-kappa B DNA binding. Conclusions These data indicate that TP inhibits ox–LDL-induced endothelial inflammation, possibly via suppression of the oxidative stress-dependent activation of the nuclear factor-kappa B signaling pathway.
Collapse
Affiliation(s)
| | | | | | - Youping Wang
- Address correspondence to: Youping Wang, MD, PhD, Central Laboratory and Division of Cardiology, First Affiliated Hospital, Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
23
|
Markers of Inflammation, Oxidative Stress, and Fibrosis in Patients with Atrial Fibrillation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4556671. [PMID: 35651726 PMCID: PMC9150993 DOI: 10.1155/2022/4556671] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 11/17/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia in clinical practice. The pathogenesis of AF is linked to inflammatory reaction and oxidative stress, which leads to fibrosis of the atria and progression of the disease. The purpose of this study was to define the role of several biomarkers of inflammation, fibrosis, and oxidative stress (OxS). We included 75 patients with paroxysmal/persistent AF, who were admitted for electrical cardioversion or pulmonary vein isolation (PVI). High-sensitivity C-reactive protein (hsCRP), galectin-3 (Gal-3), myeloperoxidase (MPO), oxidized low-density lipoprotein (oxLDL), and N-terminal pro-brain natriuretic peptide (NT-proBNP) were measured before the procedures. We compared the results with those of 75 healthy age-, sex-, and blood pressure-matched individuals. The patients were followed up for 1 year after the intervention to establish the recurrence of AF and its association with the measured markers. Patients with AF had higher MPO (52.6 vs. 36.2 ng/ml, p < 0.001) and NT-proBNP (209.0 vs. 28.0 pg/ml, p < 0.001) compared to healthy subjects. Also, they showed significantly higher levels of hsCRP (1.5 vs. 1.1 mg/l, p = 0.001) and Gal-3 (11.4 vs. 9.7 mg/l, p = 0.003), while there was no difference found in oxLDL (71.5 vs. 71.7 U/l, p = 0.449). MPO (OR = 1.012, p = 0.014), hsCRP (OR = 1.265, p = 0.026), and weight (OR = 1.029, p = 0.013) were independently associated with AF in a multivariable logistic regression analysis. Patients with successful maintenance of sinus rhythm (SR) for one year had lower baseline MPO (40.5 vs. 84.3 ng/ml, p = 0.005) and NT-proBNP (127.5 vs. 694.0 pg/ml, p < 0.001) compared to patients with recurrent AF episodes, but there was no difference in hsCRP, Gal-3, or oxLDL between them. MPO (OR = 0.985, p = 0.010) was independently associated with AF recurrence during the follow-up period when adjusted for cofounders. Patients with AF had increased markers of inflammation and fibrosis, while there was no increase detected in the OxS marker oxLDL. MPO was independently associated with AF in a multivariate model. Inflammatory and fibrotic mechanisms are important factors in electrical and structural remodelling progress in the atria of patients with AF.
Collapse
|
24
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling. Mol Med Rep 2022; 25:161. [PMID: 35293601 PMCID: PMC8941522 DOI: 10.3892/mmr.2022.12678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 11/06/2022] Open
Abstract
Endothelial cells are an important component of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Sestrin 1 (SESN1) has an important role in atherosclerosis by inhibiting NOD-like receptor family pyrin domain containing 3 inflammasome activation. However, whether SESN1 is involved in human umbilical vein endothelial cell (HUVEC) injury caused by atherosclerosis has remained to be elucidated. The present study aimed to investigate the functions of SESN1 in the inflammatory response, apoptosis and endothelial-mesenchymal transition (EndMT) of HUVECs following stimulation with oxidized low-density lipoprotein (Ox-LDL). SESN1 expression at the mRNA and protein levels was detected using reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis. Following SESN1 overexpression in Ox-LDL-stimulated HUVECs, cell viability was determined using a Cell Counting Kit-8 assay. Terminal deoxynucleotidyl transferase-mediated nick-end labeling staining was employed to detect cell apoptosis and western blot analysis was used to determine the levels of apoptosis-related proteins. RT-qPCR, ELISA and western blot were utilized to determine the levels of inflammatory factors. Immunofluorescence staining, RT-qPCR and western blot analysis were employed to assess the EndMT of Ox-LDL-stimulated HUVECs. The results revealed that SESN1 exhibited a low expression in HUVECs following Ox-LDL stimulation. SESN1 overexpression suppressed inflammation, apoptosis and EndMT in Ox-LDL-induced HUVECs. In addition, SESN1 stimulated adenosine monophosphate-activated protein kinase catalytic subunit α1/sirtuin 1 signaling to suppress Ox-LDL receptor-1 expression. An AMPK and SIRT1 inhibitor reversed the effects of SESN1 overexpression on the inflammatory response, apoptosis and EndMT of HUVECs exposed to Ox-LDL. Taken together, the present study demonstrated that SENS1 exerts a suppressive effect on Ox-LDL-induced inflammation, apoptosis and EndMT of HUVECs, suggesting that SENS1 may be used as a novel biomarker for endothelial injury-related disorders.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| |
Collapse
|
25
|
Jia J, Wang Y, Huang R, Du F, Shen X, Yang Q, Li J. Protein disulfide-isomerase A3 knockdown attenuates oxidized low-density lipoprotein-induced oxidative stress, inflammation and endothelial dysfunction in human umbilical vein endothelial cells by downregulating activating transcription factor 2. Bioengineered 2022; 13:1436-1446. [PMID: 34983301 PMCID: PMC8805980 DOI: 10.1080/21655979.2021.2018980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/11/2021] [Indexed: 11/03/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease implicated in oxidative stress and endothelial dysfunction. Protein disulfide-isomerase A3 (PDIA3) has been reported to regulate oxidative stress and suppress inflammation. This study aimed to explore the function of PDIA3 in atherosclerosis and the underlying mechanisms. PDIA3 expression in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) was detected using RT-qPCR and Western blotting. Following PDIA3 knockdown through transfection with small interfering RNA targeting PDIA3, cell viability, oxidative stress and inflammation in ox-LDL-induced HUVECs was examined using a Cell Counting Kit-8, corresponding kits and ELISA, respectively. The levels of CD31, α-smooth muscle, iNOS, p-eNOS, eNOS and NO were assessed using RT-qPCR, Western blotting and an NO kit to reflect endothelial dysfunction in ox-LDL-induced HUVECs. The relationship between PDIA3 and the activating transcription factor 2 (ATF2) was confirmed using co-immunoprecipitation. In addition, ATF2 expression was examined following PDIA3 silencing. The results indicated that PDIA3 was highly expressed in ox-LDL-induced HUVECs. PDIA3 silencing increased cell viability, and reduced oxidative stress and inflammation, as evidenced by the decreased levels of reactive oxygen species, malondialdehyde, TNF-α, IL-1β and IL-6, and increased superoxide dismutase and glutathione peroxidase activity. In addition, PDIA3 deletion improved endothelial dysfunction. PDIA3 interacted with ATF2, and PDIA3 deletion downregulated ATF2 expression. Furthermore, ATF2 overexpression reversed the effects of PDIA3 knockdown on ox-LDL-induced damage of HUVECs. Collectively, PDIA3 knockdown was found to attenuate ox-LDL-induced oxidative stress, inflammation and endothelial dysfunction in HUVECs by downregulating ATF2 expression, showing promise for the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Jia
- Department of Anesthetic Surgery, Baotou Steel Hospital, Baotou, China
| | - Yueping Wang
- Department of Cardiology, Baotou Steel Hospital, Baotou, China
| | - Ruijuan Huang
- Laser Treatment Center, Baotou Steel Hospital, Baotou, China
| | - Fengxia Du
- Department of Intensive Medicine, Baotou Steel Hospital, Baotou, China
| | - Xiaozhu Shen
- Department of Geriatrics, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Qiurong Yang
- Nursing Department, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Juan Li
- Nursing Department, The Second People’s Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
26
|
The Diagnostic and Therapeutic Potential of Galectin-3 in Cardiovascular Diseases. Biomolecules 2021; 12:biom12010046. [PMID: 35053194 PMCID: PMC8774137 DOI: 10.3390/biom12010046] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/25/2021] [Accepted: 12/27/2021] [Indexed: 12/25/2022] Open
Abstract
Galectin-3 plays a prominent role in chronic inflammation and has been implicated in the development of many disease conditions, including heart disease. Galectin-3, a regulatory protein, is elevated in both acute and chronic heart failure and is involved in the inflammatory pathway after injury leading to myocardial tissue remodelling. We discussed the potential utility of galectin-3 as a diagnostic and disease severity/prognostic biomarker in different cardio/cerebrovascular diseases, such as acute ischemic stroke, acute coronary syndromes, heart failure and arrhythmogenic cardiomyopathy. Over the last decade there has been a marked increase in the understanding the role of galectin-3 in myocardial fibrosis and inflammation and as a therapeutic target for the treatment of heart failure and myocardial infarction.
Collapse
|
27
|
Galectins in Endothelial Cell Biology and Angiogenesis: The Basics. Biomolecules 2021; 11:biom11091386. [PMID: 34572599 PMCID: PMC8464943 DOI: 10.3390/biom11091386] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/22/2023] Open
Abstract
Angiogenesis, the growth of new blood vessels out of existing vessels, is a complex and tightly regulated process. It is executed by the cells that cover the inner surface of the vasculature, i.e., the endothelial cells. During angiogenesis, these cells adopt different phenotypes, which allows them to proliferate and migrate, and to form tube-like structures that eventually result in the generation of a functional neovasculature. Multiple internal and external cues control these processes and the galectin protein family was found to be indispensable for proper execution of angiogenesis. Over the last three decades, several members of this glycan-binding protein family have been linked to endothelial cell functioning and to different steps of the angiogenesis cascade. This review provides a basic overview of our current knowledge regarding galectins in angiogenesis. It covers the main findings with regard to the endothelial expression of galectins and highlights their role in endothelial cell function and biology.
Collapse
|
28
|
Xie Q, Peng J, Guo Y, Li F. MicroRNA-33-5p inhibits cholesterol efflux in vascular endothelial cells by regulating citrate synthase and ATP-binding cassette transporter A1. BMC Cardiovasc Disord 2021; 21:433. [PMID: 34517822 PMCID: PMC8438969 DOI: 10.1186/s12872-021-02228-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 08/24/2021] [Indexed: 12/16/2022] Open
Abstract
Background A high level of total cholesterol is associated with several lipid metabolism disorders, including atherosclerosis and cardiovascular diseases. ATP-binding cassette (ABC) transporter A1 (ABCA1) and miR-33-5p play crucial roles in atherosclerosis by controlling cholesterol efflux. While citrate is a precursor metabolite for lipid and cholesterol synthesis, little is known about the association between citrate synthase (CS) and cholesterol efflux. This study investigated the role of the miR-33-5p/ABCA1/CS axis in regulating cholesterol efflux in vascular endothelial cells (VECs). Materials and methods VECs were treated with oxidized low-density lipoprotein cholesterol (ox-LDL), or pretreated with plasmids overexpressing CS, ABCA1, siRNAs against CS and ABCA1, and an miR-33-5p inhibitor. Cell apoptosis, cellular senescence-associated β-galactosidase activity, inflammation, and cholesterol efflux were detected. Results Treatment with ox-LDL decreased ABCA1 and CS levels and increased miR-33-5p expression and apoptosis in dose-dependent manners. In contrast, treatment with the miR-33-5p inhibitor and ABCA1 and CS overexpression plasmids inhibited the above-mentioned ox-LDL-induced changes. In addition, treatment with ox-LDL decreased cholesterol efflux, induced aging, and promoted the production of inflammatory cytokines (i.e., IL-6 and tumor necrosis factor TNF-α), as well as the expression of Bax and Caspase 3 proteins in VECs. All these changes were rescued by miR-33-5p inhibition and ABCA1 and CS overexpression. The inhibition of ABCA1 and CS by siRNAs eliminated the effects mediated by the miR-33-5p inhibitor, and knockdown of CS eliminated the effects of ABCA1 on VECs. Conclusions This study demonstrated the crucial roles played by the miR-33-5p/ABCA1/CS axis in regulating cholesterol efflux, inflammation, apoptosis, and aging in VECs, and also suggested the axis as a target for managing lipid metabolism disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-021-02228-7.
Collapse
Affiliation(s)
- Qiong Xie
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Jianqiang Peng
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Ying Guo
- Department of Cardiology, Hunan Provincial People's Hospital, The First Hospital Affiliated With Hunan Normal University, Changsha, 410005, Hunan, People's Republic of China
| | - Feng Li
- Departments of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, middle Ren-Min Road No. 139, Changsha, 410011, Hunan, People's Republic of China.
| |
Collapse
|
29
|
Hsu BG, Wang CH, Lai YH, Tsai JP. Serum Galectin-3 Level Is Positively Associated with Endothelial Dysfunction in Patients with Chronic Kidney Disease Stage 3 to 5. Toxins (Basel) 2021; 13:toxins13080532. [PMID: 34437403 PMCID: PMC8402460 DOI: 10.3390/toxins13080532] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022] Open
Abstract
Galectin-3, which is a novel biomarker of cardiovascular stress and related to inflammation, could predict adverse cardiovascular events. However, its relationship with endothelial function in patients with chronic kidney disease (CKD) remains inconclusive. This study aimed to investigate the association between serum galectin-3 levels and endothelial function in patients with stages 3–5 CKD. Fasting blood samples were obtained from 130 patients. Serum galectin-3 levels were determined using the enzyme-linked immunosorbent assay. The endothelial function, demonstrated as a vascular reactivity index (VRI), was measured noninvasively through digital thermal monitoring test. Then, we sorted the patients into poor, intermediate, and good vascular reactivity (VRI < 1.0, 1.0 ≤ VRI < 2.0, and VRI ≥ 2.0), accounting for 24 (18.5%), 44 (33.8%), and 62 (47.7%) patients, respectively. As the VRI decreased, the serum galectin-3 and C-reactive protein (CRP) levels significantly increased. The galectin-3 value positively correlated with the CRP value but negatively correlated with estimated glomerular filtration rate. In multivariable stepwise linear regression analysis, serum log-transformed galectin-3 level and log-transformed CRP were significantly negatively associated with VRI values. Therefore, galectin-3 together with CRP is associated with VRI values and is a potential endothelial function modulator and a valuable biomarker of endothelial dysfunction in patients with CKD.
Collapse
Affiliation(s)
- Bang-Gee Hsu
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.); (Y.-H.L.)
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Chih-Hsien Wang
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.); (Y.-H.L.)
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Yu-Hsien Lai
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.); (Y.-H.L.)
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan; (B.-G.H.); (C.-H.W.); (Y.-H.L.)
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
- Correspondence:
| |
Collapse
|
30
|
Karsiyaka Hendek M, Olgun E, Kisa U. The effect of initial periodontal treatment on gingival crevicular fluid galectin-3 levels in participants with periodontal disease. Aust Dent J 2021; 66:169-174. [PMID: 33378559 DOI: 10.1111/adj.12815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND The aim of study was to evaluate galectin-3 levels in gingival crevicular fluid (GCF) from periodontally healthy (H) patients and those with periodontitis (P), gingivitis (G) and the effect of initial periodontal treatment on GCF galectin-3 level. METHODS A total of 75 participants, 25 patients with periodontitis, 25 with gingivitis and 25 periodontally healthy subjects were included into the study. Patients with periodontal disease received initial periodontal treatment. GCF galectin-3 level was assessed at baseline and at the 6th-8th weeks after completion of periodontal treatment. GCF galectin-3 level was evaluated by enzyme-linked immunosorbent assay. RESULTS GCF galectin-3 level was the lowest in the H group (102.31[63.07] μg/30 s), followed by the G group (241.45[145.89] μg/30 s) and the highest in the P group (338.27[219.37] μg/30 s). These differences were statistically significant between H and the other groups (P < 0.001). After initial periodontal treatment, GCF galectin-3 level significantly decreased in the G and P groups compared to baseline values (P < 0.01). CONCLUSION The results of this study suggest that GCF galectin-3 level is a potential biomarker for the evaluation of gingival inflammation and initial periodontal treatment is effective in decreasing GCF galectin-3 level.
Collapse
Affiliation(s)
- M Karsiyaka Hendek
- Department of Periodontology, Faculty of Dentistry, Kirikkale University, Kirikkale, Turkey
| | - E Olgun
- Department of Periodontology, Faculty of Dentistry, Kirikkale University, Kirikkale, Turkey
| | - U Kisa
- Department of Biochemistry, Faculty of Medicine, Kirikkale University, Kirikkale, Turkey
| |
Collapse
|
31
|
Malaviya R, Abramova EV, Rancourt RC, Sunil VR, Napierala M, Weinstock D, Croutch CR, Roseman J, Tuttle R, Peters E, Casillas RP, Laskin JD, Laskin DL. Progressive Lung Injury, Inflammation, and Fibrosis in Rats Following Inhalation of Sulfur Mustard. Toxicol Sci 2020; 178:358-374. [PMID: 33002157 PMCID: PMC7751178 DOI: 10.1093/toxsci/kfaa150] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Sulfur mustard (SM) inhalation causes debilitating pulmonary injury in humans which progresses to fibrosis. Herein, we developed a rat model of SM toxicity which parallels pathological changes in the respiratory tract observed in humans. SM vapor inhalation caused dose (0.2-0.6 mg/kg)-related damage to the respiratory tract within 3 days of exposure. At 0.4-0.6 mg/kg, ulceration of the proximal bronchioles, edema and inflammation were observed, along with a proteinaceous exudate containing inflammatory cells in alveolar regions. Time course studies revealed that the pathologic response was biphasic. Thus, changes observed at 3 days post-SM were reduced at 7-16 days; this was followed by more robust aberrations at 28 days, including epithelial necrosis and hyperplasia in the distal bronchioles, thickened alveolar walls, enlarged vacuolated macrophages, and interstitial fibrosis. Histopathologic changes were correlated with biphasic increases in bronchoalveolar lavage (BAL) cell and protein content and proliferating cell nuclear antigen expression. Proinflammatory proteins receptor for advanced glycation end product (RAGE), high-mobility group box protein (HMGB)-1, and matrix metalloproteinase (MMP)-9 also increased in a biphasic manner following SM inhalation, along with surfactant protein-D (SP-D). Tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS), inflammatory proteins implicated in mustard lung toxicity, and the proinflammatory/profibrotic protein, galectin (Gal)-3, were upregulated in alveolar macrophages and in bronchiolar regions at 3 and 28 days post-SM. Inflammatory changes in the lung were associated with oxidative stress, as reflected by increased expression of heme oxygenase (HO)-1. These data demonstrate a similar pathologic response to inhaled SM in rats and humans suggesting that this rodent model can be used for mechanistic studies and for the identification of efficacious therapeutics for mitigating toxicity.
Collapse
Affiliation(s)
- Rama Malaviya
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Raymond C Rancourt
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Vasanthi R Sunil
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854
| | - Marta Napierala
- Laboratory of Environmental Research, Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Daniel Weinstock
- Janssen Boitherapeutics, Janssen Research & Development, Spring House, Pennsylvania 19477
| | - Claire R Croutch
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Julie Roseman
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Rick Tuttle
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | - Eric Peters
- Medical Research Portfolio, MRIGlobal, Kansas City, Missouri 64110
| | | | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey 08854
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey 08854,To whom correspondence should be addressed at Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, 160 Frelinghuysen Road, Piscataway, NJ 08854. E-mail:
| |
Collapse
|
32
|
Improvement of Endothelial Dysfunction of Berberine in Atherosclerotic Mice and Mechanism Exploring through TMT-Based Proteomics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8683404. [PMID: 32566106 PMCID: PMC7284929 DOI: 10.1155/2020/8683404] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a multifactorial vascular disease triggered by disordered lipid metabolism, characterized by chronic inflammatory injury, and initiated by endothelial dysfunction. Berberine is the main active alkaloid of the herbal medicine Coptidis Rhizoma (Huanglian). Notably, berberine has been shown to have beneficial effects against atherosclerosis. However, the mechanisms of berberine in preventing atherosclerosis are still unclear. This study is aimed at investigating the effects and mechanisms of berberine in protecting the aorta and ameliorating atherosclerosis in apolipoprotein E-deficient (ApoE−/−) mice. Here, we demonstrated that berberine reduced serum lipid levels, antagonized hepatic lipid accumulation, improved intima-media thickening, and alleviated atherosclerotic lesions in ApoE−/− mice fed a western-type diet for 12 weeks. Meanwhile, berberine reduced aortic reactive oxygen species (ROS) generation and reduced the serum levels of malondialdehyde (MDA), oxidized low-density lipoprotein (ox-LDL), and interleukin-6 (IL-6). In aortic ring assay, berberine restored aortic endothelium-dependent vasodilatation in vivo and in vitro. Furthermore, 4,956 proteins were identified by proteomic analysis, and 199 differentially expressed proteins regulated by berberine were found to be involved in many biological pathways, such as mitochondrial dysfunction, fatty acid β-oxidation I, and FXR/RXR activation. Summarily, these data suggested that berberine ameliorates endothelial dysfunction and protects against atherosclerosis, and thus may be a promising therapeutic candidate for atherosclerosis.
Collapse
|
33
|
Wang JY, Lu PH, Lin WW, Wei YH, Chiu LY, Chern SR, Hung CF, Wu NL. Galectin-3 regulates UVB-induced inflammation in skin. J Dermatol Sci 2020; 98:119-127. [PMID: 32312639 DOI: 10.1016/j.jdermsci.2020.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Galectin-3 is widely expressed in many immunocytes and epithelial cells including skin keratinocytes. Galectin-3 can regulate immunological or inflammatory processes and plays a proinflammatory role in some disease models. Galectin-3 has a role in disorders related to ultraviolet (UV) photodamage such as apoptosis, skin squamous cell carcinoma and basal cell carcinoma. However, the evidence of galectin-3 in UVB-induced skin inflammation is still limited and the underlying molecular mechanism remains elusive. OBJECTIVE We aimed to investigate the effects of galectin-3 in human epidermal keratinocytes and in mice after UVB irradiation. METHODS Primary human epidermal keratinocytes with galectin-3 knockdown were used as the in vitro model. ELISA, QPCR, and western blotting were applied to evaluate the released cytokine, mRNA and protein expression. Histologic analysis, measurement of erythema and transepidermal water loss (TEWL) were applied to evaluate UVB-induced skin damage in galectin-3 knockout mice. RESULTS In UVB-irradiated human keratinocytes, galectin-3 knockdown downregulated the UVB-induced ASC crosslinking, cleavage of caspase-1, and formation of active IL-1β. Galectin-3 knockdown also decreased UVB-induced production of reactive oxygen species, p38 phosphorylation, and COX2 expression in human keratinocytes. After four days of UVB irradiation, galectin-3 knockout mice showed reduced gross erythema, histologic features of tissue inflammation, quantified levels of erythema and TEWL compared to wild type mice. The skin tissue lysate also showed less expression of active IL-1β and COX2 in galectin-3 knockout mice. CONCLUSION Galectin-3 may play a positive regulatory role in UVB-induced skin inflammation.
Collapse
Affiliation(s)
- Jen-Yu Wang
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan.
| | - Po-Hsuan Lu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yu-Hsuan Wei
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Ling-Ya Chiu
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Schu-Rern Chern
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| | - Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan.
| |
Collapse
|
34
|
Galectin-3 Is a Potential Mediator for Atherosclerosis. J Immunol Res 2020; 2020:5284728. [PMID: 32149158 PMCID: PMC7042544 DOI: 10.1155/2020/5284728] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/13/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic inflammatory arterial disease forming the pathological basis of many cardiovascular diseases such as coronary heart disease, heart failure, and stroke. Numerous studies have implicated inflammation as a key player in the initiation and progression of atherosclerosis. Galectin-3 (Gal-3) is a 30 kDa β-galactose, highly conserved and widely distributed intracellularly and extracellularly. Gal-3 has been demonstrated in recent years to be a novel inflammatory factor participating in the process of intravascular inflammation, lipid endocytosis, macrophage activation, cellular proliferation, monocyte chemotaxis, and cell adhesion. This review focuses on the role of Gal-3 in atherosclerosis and the mechanism involved and several classical Gal-3 agonists and antagonists in the current studies.
Collapse
|
35
|
Naringin Reverses High-Cholesterol Diet-Induced Vascular Dysfunction and Oxidative Stress in Rats via Regulating LOX-1 and NADPH Oxidase Subunit Expression. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3708497. [PMID: 31781614 PMCID: PMC6855071 DOI: 10.1155/2019/3708497] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/10/2019] [Indexed: 12/29/2022]
Abstract
Hypercholesterolaemia is associated with oxidative stress and endothelial dysfunction and leads to the development of atherosclerosis. Naringin exhibits cardiovascular protective and antioxidant properties. Therefore, the aim of this study was to assess the effect of naringin administration on vascular oxidative stress and endothelial dysfunction in hypercholesterolaemic rats and to elucidate its underlying mechanism. Sprague Dawley rats were fed a diet with 1.5% cholesterol (HCD) for 8 weeks to induce hypercholesterolaemia. Naringin (100 mg/kg body weight) was orally administrated to rats during the last 4 weeks of the diet treatment. After 8 weeks, the thoracic aorta was isolated to determine vascular function and nitric oxide (NO) levels. The aortic superoxide anion (O2−) level was detected using dihydroethidium (DHE) fluorescence staining. Protein expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits, and inducible nitric oxide synthase (iNOS), as well as oxidative damage markers, was also evaluated in aortae. Naringin treatment of hypercholesterolaemic rats enhanced aortic NO levels, restored endothelium-dependent responses to acetylcholine (ACh), and reduced aortic O2− levels. Furthermore, naringin treatment decreased LOX-1, NADPH oxidase subunits (p47phox, Nox2, and Nox4), and iNOS as well as oxidative damage markers (3-nitrotyrosine (3-NT) and 4-hydroxynonenal (4-HNE)) expression in aortic tissues from hypercholesterolaemic rats. These results demonstrate that naringin treatment improves endothelium dysfunction in hypercholesterolaemic rats, at least partially by decreasing oxidative stress via downregulation of LOX-1 and NADPH oxidase.
Collapse
|
36
|
PKR Promotes Oxidative Stress and Apoptosis of Human Articular Chondrocytes by Causing Mitochondrial Dysfunction through p38 MAPK Activation-PKR Activation Causes Apoptosis in Human Chondrocytes. ANTIOXIDANTS (BASEL, SWITZERLAND) 2019. [PMID: 31484360 DOI: 10.3390/antiox8090370.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Osteoarthritis (OA) is one of the most common types of arthritis in the elderly people. It has been known that chondrocyte apoptosis occurs in OA cartilage; however, the detailed molecular mechanism remains unclear. In the current study, we aimed to elucidate the role of double-stranded RNA-dependent protein kinase R (PKR) in the TNF-α-caused apoptosis in chondrocytes. Human articular chondrocytes were digested from cartilages of OA subjects who accepted arthroplastic knee surgery. Our results showed that phosphorylation of p38 MAPK was increased after TNF-α stimulation or PKR activation using poly (I:C), and TNF-α-induced p38 MAPK upregulation was inhibited by PKR inhibition, suggesting phosphor-p38 MAPK was regulated by PKR. Moreover, we found that PKR participated in the p53-dependent destruction of AKT following activation of p38 MAPK. The inhibition of AKT led to the reduced expression of PGC-1α, which resulted in mitochondrial dysfunction and increased oxidative stress. We showed that the reduction of oxidative stress using antioxidant Mito TEMPO lowered the TNF-α-induced caspase-3 activation and TUNEL-positive apoptotic cells. The diminished apoptotic response was also observed after repression of PKR/p38 MAPK/p53/AKT/PGC-1α signaling. Taken together, we demonstrated that the aberrant mitochondrial biogenesis and increased oxidative stress in chondrocytes after TNF-α stimulation were mediated by PKR, which may contribute to the chondrocyte apoptosis and cartilage degeneration in OA.
Collapse
|
37
|
PKR Promotes Oxidative Stress and Apoptosis of Human Articular Chondrocytes by Causing Mitochondrial Dysfunction through p38 MAPK Activation-PKR Activation Causes Apoptosis in Human Chondrocytes. Antioxidants (Basel) 2019; 8:antiox8090370. [PMID: 31484360 PMCID: PMC6769915 DOI: 10.3390/antiox8090370] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is one of the most common types of arthritis in the elderly people. It has been known that chondrocyte apoptosis occurs in OA cartilage; however, the detailed molecular mechanism remains unclear. In the current study, we aimed to elucidate the role of double-stranded RNA-dependent protein kinase R (PKR) in the TNF-α-caused apoptosis in chondrocytes. Human articular chondrocytes were digested from cartilages of OA subjects who accepted arthroplastic knee surgery. Our results showed that phosphorylation of p38 MAPK was increased after TNF-α stimulation or PKR activation using poly (I:C), and TNF-α-induced p38 MAPK upregulation was inhibited by PKR inhibition, suggesting phosphor-p38 MAPK was regulated by PKR. Moreover, we found that PKR participated in the p53-dependent destruction of AKT following activation of p38 MAPK. The inhibition of AKT led to the reduced expression of PGC-1α, which resulted in mitochondrial dysfunction and increased oxidative stress. We showed that the reduction of oxidative stress using antioxidant Mito TEMPO lowered the TNF-α-induced caspase-3 activation and TUNEL-positive apoptotic cells. The diminished apoptotic response was also observed after repression of PKR/p38 MAPK/p53/AKT/PGC-1α signaling. Taken together, we demonstrated that the aberrant mitochondrial biogenesis and increased oxidative stress in chondrocytes after TNF-α stimulation were mediated by PKR, which may contribute to the chondrocyte apoptosis and cartilage degeneration in OA.
Collapse
|