1
|
Khasanov R, Svoboda D, Tapia-Laliena MÁ, Kohl M, Maas-Omlor S, Hagl CI, Wessel LM, Schäfer KH. Muscle hypertrophy and neuroplasticity in the small bowel in short bowel syndrome. Histochem Cell Biol 2023; 160:391-405. [PMID: 37395792 PMCID: PMC10624713 DOI: 10.1007/s00418-023-02214-4] [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] [Accepted: 05/24/2023] [Indexed: 07/04/2023]
Abstract
Short bowel syndrome (SBS) is a severe, life-threatening condition and one of the leading causes of intestinal failure in children. Here we were interested in changes in muscle layers and especially in the myenteric plexus of the enteric nervous system (ENS) of the small bowel in the context of intestinal adaptation. Twelve rats underwent a massive resection of the small intestine to induce SBS. Sham laparotomy without small bowel transection was performed in 10 rats. Two weeks after surgery, the remaining jejunum and ileum were harvested and studied. Samples of human small bowel were obtained from patients who underwent resection of small bowel segments due to a medical indication. Morphological changes in the muscle layers and the expression of nestin, a marker for neuronal plasticity, were studied. Following SBS, muscle tissue increases significantly in both parts of the small bowel, i.e., jejunum and ileum. The leading pathophysiological mechanism of these changes is hypertrophy. Additionally, we observed an increased nestin expression in the myenteric plexus in the remaining bowel with SBS. Our human data also showed that in patients with SBS, the proportion of stem cells in the myenteric plexus had risen by more than twofold. Our findings suggest that the ENS is tightly connected to changes in intestinal muscle layers and is critically involved in the process of intestinal adaptation to SBS.
Collapse
Affiliation(s)
- Rasul Khasanov
- Department of Pediatric Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Daniel Svoboda
- Department of Pediatric Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - María Ángeles Tapia-Laliena
- Department of Pediatric Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Martina Kohl
- Department of Pediatric and Adolescent Medicine, University Medical Center Schleswig-Holstein, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Silke Maas-Omlor
- Enteric Nervous System Group, University of Applied Sciences Kaiserslautern, Amerikastrasse 1, 66482, Zweibrücken, Germany
| | - Cornelia Irene Hagl
- Carl Remigius Medical School, Charles de Gaulle Str. 2, 81737, Munich, Germany
| | - Lucas M Wessel
- Department of Pediatric Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Karl-Herbert Schäfer
- Enteric Nervous System Group, University of Applied Sciences Kaiserslautern, Amerikastrasse 1, 66482, Zweibrücken, Germany
| |
Collapse
|
2
|
Yang P, Zang G, Yan Y, Zhong W, Li B, Xu Y, Shao C, Wang Z, Pu J, Yuan W. CD137-CD137L Aggravates Calcification of Vascular Smooth Muscle Cell and Vasculature of ApoE -/- Mice Via Rab7-Mediated Autophagy. J Cardiovasc Transl Res 2022; 15:1297-1314. [PMID: 35763154 DOI: 10.1007/s12265-022-10272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Vascular calcification is an independent risk factor for acute cardiovascular events and a predictor of adverse prognosis; the abnormal fusion and degradation of autophagosomes and lysosomes are closely related to the calcification of VSMC and aortic AS plaque in ApoE-/- mice. Rab7 is a member of the Ras protein family and acts as a molecular switch in the fusion between autophagosomes and lysosomes. In this study, we found that the activation of the CD137-CD137L signal promoted calcification by inhibiting the expression and activity of Rab7, which regulates the degradation of autophagic cargo in vascular smooth muscle cells (VSMCs) and aortic atherosclerosis (AS) plaques in ApoE-/- mice. Knockdown of Rab7 impaired its tethering with the downstream molecule FYVE and coiled-coil containing 1 (FYCO1), which transports autophagosomes to lysosomes through microtubule motor kinesins and fuses with lysosomes to degrade the autophagic content. Overexpression of Rab7-alleviated calcification caused by the activation of the CD137 signaling pathway. In addition, FYCO1 knockdown promoted calcification even though the expression and activity of Rab7 were normal. Our results suggest that Rab7 is the target of CD137 signaling; Rab7 cannot interact with its downstream molecule FYCO1 when its activity and expression were inhibited by the activation of CD137 signaling pathway, thus inhibiting the autophagic degradation and promoting calcification.
Collapse
Affiliation(s)
- Ping Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Guangyao Zang
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Yang Yan
- Department of Cardiology Ren Ji Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhong
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Bo Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Yao Xu
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Jun Pu
- Department of Cardiology Ren Ji Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China.
| |
Collapse
|
3
|
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: 6] [Impact Index Per Article: 3.0] [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
|
4
|
Association between Endothelial Cell-Specific Molecule 1 and Galectin-3 in Patients with ST-Segment Elevation Myocardial Infarction: A Pilot Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1723309. [PMID: 36388167 PMCID: PMC9646309 DOI: 10.1155/2022/1723309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/22/2022] [Indexed: 11/07/2022]
Abstract
The biomarkers galectin-3 (Gal-3) and endothelial cell-specific molecule 1 (ESM-1) reflect endothelial function and inflammation. As a consequence, they play an important role in both the diagnosis and characterization of ST-segment elevation myocardial infarction (STEMI). However, no prior study has explored the association between ESM-1 and Gal-3 in STEMI patients. This study is aimed at determining the ESM-1 and Gal-3 levels in the serum of STEMI patients and then exploring the correlation between the levels of these two biomarkers and their clinical significance in STEMI patients. The participants were divided into two groups: the ST group comprised 35 hospitalized STEMI patients while the control group comprised 24 people with normal coronary arteries. In all the patients, venous blood was taken from the middle of the antecubital fossa. The serum ESM-1 and Gal-3 concentrations were determined using an enzyme-linked immunosorbent assay. The results revealed that the ESM-1 and Gal-3 levels in the STEMI patients were 1.6 and 2.8 times higher, respectively, when compared with the controls (P < 0.001). Moreover, the ESM-1 and Gal-3 levels exhibited a positive linear correlation (r = 0.758, P < 0.001) in the acute STEMI patients. In conclusion, the ESM-1 and Gal-3 levels were found to be significantly elevated and correlated in the STEMI patients. Thus, combining these two biomarkers of endothelial dysfunction and inflammation might be useful for the diagnosis and assessment of STEMI.
Collapse
|
5
|
Tian L, Wang Y, Zhang R. Galectin-3 induces vascular smooth muscle cells calcification via AMPK/TXNIP pathway. Aging (Albany NY) 2022; 14:5086-5096. [PMID: 35771146 PMCID: PMC9271303 DOI: 10.18632/aging.204130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 05/23/2022] [Indexed: 12/15/2022]
Abstract
Galectin-3 plays an important role in atherosclerosis. Upregulation of VSMCs calcification is involved in the progression and development of vulnerable plaques. Thioredoxin-interacting protein (TXNIP) has been regarded as an important determinant in regulating inflammation and oxidative stress. In this study, we evaluated the role of TXNIP in galectin-3-induced vascular calcification. A primary culture of mouse VSMCs was established by enzymatic digestion of aorta. Small interfering (si) RNA was used to knock down the expression of target gene. VSMCs were treated with 3-methyladenine (3-MA) or compound C respectively. Western blot was performed to detect the protein level in VSMCs, Alkaline phosphatase (ALP) and Alizarin red staining was used to observe calcium deposition. Dihydroethidium (DHE) staining was used to observe the reactive oxygen species (ROS) production. Here we showed that galectin-3 increased aorta and VSMCs calcification, which was associated with AMPK/TXNIP upregulation and autophagy activation. TXNIP inhibition decreased galectin-3-induced aorta and VSMCs calcification and autophagy activation. 3-MA or Atg5 siRNA decreased galectin-3-induced upregulation of Runx2, BMP2 and OPN. AMPK mediated galectin-3-induced VSMCs osteogenic differentiation. These findings illustrated that TXNIP mediated galectin-3-induced vascular calcification, AMPK and autophagy activation were also associated with this process.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Li J, Shen H, Owens GK, Guo LW. SREBP1 regulates Lgals3 activation in response to cholesterol loading. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:892-909. [PMID: 35694209 PMCID: PMC9168384 DOI: 10.1016/j.omtn.2022.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 05/12/2022] [Indexed: 12/02/2022]
Abstract
Aberrant smooth muscle cell (SMC) plasticity is etiological to vascular diseases. Cholesterol induces SMC phenotypic transition featuring high LGALS3 (galectin-3) expression. This proatherogenic process is poorly understood for its molecular underpinnings, in particular, the mechanistic role of sterol regulatory-element binding protein-1 (SREBP1), a master regulator of lipid metabolism. Herein we show that cholesterol loading stimulated SREBP1 expression in mouse, rat, and human SMCs. SREBP1 positively regulated LGALS3 expression (and vice versa), whereas Krüppel-like factor-15 (KLF15) acted as a negative regulator. Both bound to the Lgals3 promoter, yet at discrete sites, as revealed by chromatin immunoprecipitation-qPCR and electrophoretic mobility shift assays. SREBP1 and LGALS3 each abated KLF15 protein, and blocking the bromo/extraterminal domain-containing proteins (BETs) family of acetyl-histone readers abolished cholesterol-stimulated SREBP1/LGALS3 protein production. Furthermore, silencing bromodomain protein 2 (BRD2; but not other BETs) reduced SREBP1; endogenous BRD2 co-immunoprecipitated with SREBP1's transcription-active domain, its own promoter DNA, and that of L gals 3. Thus, results identify a previously uncharacterized cholesterol-responsive dyad-SREBP1 and LGALS3, constituting a feedforward circuit that can be blocked by BETs inhibition. This study provides new insights into SMC phenotypic transition and potential interventional targets.
Collapse
Affiliation(s)
- Jing Li
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Hongtao Shen
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Gary K. Owens
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
7
|
Tian L, Ding F, Zhang R. Predictive Value of Galectin-3 and Brachial-Ankle Pulse Wave Velocity for Coronary Artery Calcification in Coronary Arteriography Patients. Int J Clin Pract 2022; 2022:1865736. [PMID: 35685493 PMCID: PMC9159218 DOI: 10.1155/2022/1865736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES To study the predictive value for coronary artery calcification (CAC) of plasma galectin-3 and brachial-ankle pulse wave velocity (BaPWV) in coronary arteriography (CAG) patients. METHODS Patients who received coronary arteriography (CAG) examination were recruited. The level of plasma galectin-3 was measured by the enzyme-linked immunosorbent assay. The arterial stiffness was analyzed by BaPWV and ankle-brachial index (ABI) which were measured using a volume-plethysmographic device. Receiver operating characteristic (ROC) curve was used to analyze the prognostic value of galectin-3 or BaPWV for coronary artery calcification (CAC). RESULTS The level of galectin-3 and BaPWV was significantly higher in CAC patients compared with that in control (p < 0.01). The level of plasma galectin-3 was positively correlated with BaPWV (r = -0.296, p < 0.01) and negatively correlated with ABI (r = -0.296, p < 0.01). ROC curve analysis revealed that galectin-3 ≥5.90 ng/ml was the most powerful predictor for CAC with sensitivity of 85.5% and specificity of 83.5%. The area under the curve (AUC) was 0.916. When the level of BaPWV was more than 1909 m/s, the sensitivity and specificity were 61.8% and 69.6%, respectively, for predicting CAC. The AUC was 0.646. CONCLUSIONS The level of plasma galectin-3 increases significantly in CAC patients compared to control, and its level is related to BaPWV and ABI. Galectin-3 and BaPWV can be used to predict CAC, and the diagnosis value (sensitivity and specificity) of galectin-3 for CAC is better than that of BaPWV.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
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: 25] [Impact Index Per Article: 8.3] [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
|
9
|
Tian L, Huang CK, Ding F, Zhang R. Galectin-3 Mediates Thrombin-Induced Vascular Smooth Muscle Cell Migration. Front Cardiovasc Med 2021; 8:686200. [PMID: 34746246 PMCID: PMC8563778 DOI: 10.3389/fcvm.2021.686200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) migration is an important step in the progression and development of vulnerable plaques. Thrombin is involved in both physiological and pathological processes of atherosclerosis. Therefore, the elucidation of the mechanisms underlying thrombin-induced VSMC migration is essential for devising effective treatments aimed at the prevention of plaque instability. In this study, we found that thrombin activated MAPK signaling pathways and increased the expression of galectin-3, which was also a well-known factor in atherosclerosis. Knockdown of galectin-3 by specific small interfering RNA (siRNA) blocked thrombin-induced activation of ERK1/2 and p38 MAPK, but not JNK MAPK. Src/FAK phosphorylation was also shown to be activated by thrombin. FAK autophosphorylation at Y397 was most significantly inhibited by galectin-3 siRNA. Galectin-3 siRNA or specific inhibitor (P38 MAPK inhibitor and ERK1/2 inhibitor) effectively prevented thrombin-induced VSMC migration via reducing paxillin expression. These findings demonstrate, for the first time, that thrombin stimulation of VSMC migration and paxillin expression are regulated by galectin-3, and ERK1/2, p38 MAPK, and Src/FAK signaling pathways are involved in this process. These results are beneficial to clarify the role of galectin-3 in thrombin-induced advanced lesions in atherosclerosis and shed new insights into the regulatory mechanism of VSMC migration in combating plaque rupture.
Collapse
Affiliation(s)
- Lei Tian
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Kai Huang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Ibrahim BA, Mohamed SH, Hassaan MMM, Sabbah NA. Associations of galectin-3 expression and LGALS-3 (rs4652) gene variant with coronary artery disease risk in diabetics. J Med Biochem 2021; 40:395-406. [PMID: 34616230 PMCID: PMC8451226 DOI: 10.5937/jomb0-30424] [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: 01/19/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022] Open
Abstract
Background Galectin-3 protein encoded by lectin galactoside-binding soluble-3 (LGALS-3) gene is an important genetic factor in type 2 diabetes mellitus (T2DM) and its cardiovascular obstacles in various populations. We aimed to elicit the pro-inflammatory effect of galectin-3 as determined by interleukin-6 (IL-6) serum levels and to explore the relationship between galectin-3 (LGALS-3 rs4652) gene variant and its expression levels with coronary artery disease (CAD) risk among T2DM Egyptian patients. Methods 112 lean subjects were compared to 100 T2DM without CAD and 84 T2DM with CAD. A tetra-primer amplification refractory mutation system polymerase chain reaction was used to test LGALS-3 (rs4652), and galectin3 expression was tested with a quantitative real-time polymerase chain reaction. Serum IL-6 was measured using an enzyme-linked immunosorbent assay. Results We found that the prevalence of LGALS-3 (rs4652) AC genotype and galectin-3 gene expression levels in T2DM with CAD were significantly higher than the additional 2 groups and were correlated positively to IL-6 circulating levels. Also, the C allele carriers (AC+CC) had significantly higher relative Galectin-3 expression levels compared to the A allele carriers (AA). Conclusions We concluded that galectin-3 expression levels and LGALS-3 (rs4652) AC genotype were coronary artery disease risk factors in people with type two diabetes among an Egyptian sample.
Collapse
Affiliation(s)
- Basma A Ibrahim
- University of Zagazig, Faculty of Medicine, Medical Biochemistry Department, Zagazig, Egypt
| | - Samy H Mohamed
- University of Zagazig, Faculty of Medicine, Medical Biochemistry Department, Zagazig, Egypt
| | - Mohamed M M Hassaan
- University of Zagazig, Faculty of Medicine, Internal Medicine Department, Zagazig, Egypt
| | - Norhan A Sabbah
- University of Zagazig, Faculty of Medicine, Medical Biochemistry Department, Zagazig, Egypt
| |
Collapse
|
11
|
Cheng ZB, Huang L, Xiao X, Sun JX, Zou ZK, Jiang JF, Lu C, Zhang HY, Zhang C. Irisin in atherosclerosis. Clin Chim Acta 2021; 522:158-166. [PMID: 34425103 DOI: 10.1016/j.cca.2021.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Irisin, a novel exercise-induced myokine, has been shown to play important roles in increasing white adipose tissue browning, regulating energy metabolism and improving insulin resistance. Growing evidence suggests a direct role for irisin in preventing atherosclerosis (AS) by inhibiting oxidative stress, improving dyslipidemia, facilitating anti-inflammation, reducing cellular damage and recovering endothelial function. In addition, some studies have noted that serum irisin levels play an essential role in cardiovascular diseases (CVDs) risk prediction, highlighting that irisin has the potential to be a useful predictive marker and therapeutic target of AS, especially in monitoring therapeutic efficacy. This review summarizes the understanding of irisin-mediated regulation in essential biological pathways and functions in atherosclerosis and prompts further exploitation of the biological properties of irisin in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Zhe-Bin Cheng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Jia-Xiang Sun
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Zi-Kai Zou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jie-Feng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Lu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hai-Ya Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
12
|
Obeid S, Yousif N, Davies A, Loretz R, Saleh L, Niederseer D, Noor HA, Amin H, Mach F, Gencer B, Räber L, Windecker S, Templin C, Nanchen D, Rodondi N, Muller O, Matter CM, von Eckardstein A, Lüscher TF. Prognostic role of plasma galectin-3 levels in acute coronary syndrome. EUROPEAN HEART JOURNAL-ACUTE CARDIOVASCULAR CARE 2020; 9:869-878. [DOI: 10.1177/2048872620974612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aim:
Cystatin C, neutrophil gelatinase-associated lipocalin and galectin-3 have emerged as biomarker candidates to predict cardiovascular outcomes and mortality in the general population as well as in patients with coronary artery or renal disease. However, their predictive role and clinical utility in patients with acute coronary syndromes alone or in combination beyond currently used risk scores remains to be determined.
Methods and results:
Cystatin C, neutrophil gelatinase-associated lipocalin, and galectin-3 were measured in plasmas of 1832 patients at the time of presentation with acute coronary syndromes requiring percutaneous coronary intervention or coronary artery bypass grafting. The primary outcomes were major adverse cardiac and cerebrovascular events (defined as the composite of all-cause mortality, cerebrovascular events, any repeat revascularization or myocardial infarction) and all-cause mortality after 1 year and occurred in 192 (10.5%) and 78 (4.3%) of patients, respectively. All three biomarkers were increased in those with major adverse cardiac and cerebrovascular events compared with those without (p<0.001). However, only galectin-3 (all-cause mortality: hazard ratio=1.027 (95% confidence interval (1.011–1.043); p=0.001), major adverse cardiac and cerebrovascular events: hazard ratio=1.025 (95% confidence interval (1.012–1.037); p<0.001)) but not cystatin C nor neutrophil gelatinase-associated lipocalin emerged as independent predictors of both major adverse cardiac and cerebrovascular events and death. The risks were particularly high in the highest quartile of galectin-3. The integration of galectin-3 into the global registry of acute coronary events (GRACE) score improved the prediction of major adverse cardiac and cerebrovascular events and all-cause mortality significantly. The areas under the receiver operator characteristics curves increased from 0.6701 to 0.6932 for major adverse cardiac and cerebrovascular events (p=0.0474) and from 0.804 to 0.8199 for all-cause mortality (p=0.0197). Finally, we applied net reclassification improvement index using different cut-offs for major adverse cardiac and cerebrovascular events which showed negative results (for the cut-offs of 5% and 15%, net reclassification improvement index 0.028, p=0.586, for the cut-offs of 10% and 20%, net reclassification improvement index 0.072, p=0.1132 and for the cut-offs of 10% and 30% the net reclassification improvement index is 0.0843, p=0.077).
Conclusion:
In acute coronary syndromes patients, galectin-3 has moderate prognostic accuracy, provides statistically significant incremental value in some, but not all models, and that the magnitude of any improvement would seem of questionable clinical value.
Collapse
Affiliation(s)
- Slayman Obeid
- Department of Cardiology, University Hospital Zurich, Switzerland
| | - Nooraldaem Yousif
- Department of Cardiology, University Hospital Zurich, Switzerland
- Mohammed Bin Khalifa Cardiac Centre, Kingdom of Bahrain
| | - Allan Davies
- Royal Brompton and Harefield Hospitals and Imperial College, UK
| | - Ruben Loretz
- Department of Cardiology, University Hospital Zurich, Switzerland
| | - Lanja Saleh
- Institute of Clinical Chemistry, University Hospital Zurich, Switzerland
| | - David Niederseer
- Department of Cardiology, University Hospital Zurich, Switzerland
| | - Husam A Noor
- Mohammed Bin Khalifa Cardiac Centre, Kingdom of Bahrain
| | - Haitham Amin
- Mohammed Bin Khalifa Cardiac Centre, Kingdom of Bahrain
| | - François Mach
- Department of Cardiology, Hospital Universitaire de Geneve, Switzerland
| | - Baris Gencer
- Department of Cardiology, Hospital Universitaire de Geneve, Switzerland
| | - Lorenz Räber
- Department of Cardiology, University Hospital Bern, Switzerland
| | | | | | - David Nanchen
- Department of Ambulatory Care and Community Medicine, University of Lausanne, Switzerland
| | - Nicolas Rodondi
- Department of General Medicine, University Hospital Bern, Switzerland
- Institute of Primary Health Care (BIHAM), University of Bern, Switzerland
| | - Olivier Muller
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois, Switzerland
| | - Christian M Matter
- Department of Cardiology, University Hospital Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Switzerland
| | | | - Thomas F Lüscher
- Royal Brompton and Harefield Hospitals and Imperial College, UK
- Center for Molecular Cardiology, University of Zurich, Switzerland
| |
Collapse
|
13
|
Long X, You G, Wu Q, Zhou Y, Yu F, Xiao Y, Deng S, Song F, Huang J, Tian M. Abnormal expression of homeobox c6 in the atherosclerotic aorta and its effect on proliferation and migration of rat vascular smooth muscle cells. Acta Biochim Biophys Sin (Shanghai) 2020; 52:935-943. [PMID: 32785574 DOI: 10.1093/abbs/gmaa080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Indexed: 11/14/2022] Open
Abstract
Homeobox c6 (Hoxc6) affects the proliferation, migration, and infiltration of malignant tumor cells; however, the effect of Hoxc6 on atherosclerosis (AS) as well as the proliferation and migration of vascular smooth muscle cells (VSMCs), which play a role in promoting AS, has not yet been well clarified. In the present study, we tested the hypothesis that Hoxc6 affects the proliferation and migration of rat VSMCs, and hence is involved in AS. The results showed that the expression of Hoxc6 mRNA and protein was higher in normal rat aortic wall than in the myocardium. Subsequently, a rat model of AS was established by high-fat feeding for 2 months. The expression of Hoxc6 mRNA and protein was increased significantly in AS lesions, while the expression of p53 protein was decreased and that of proliferating cell nuclear antigen (PCNA) was increased. Moreover, not only the proliferation and mobility of cells in normal culture were decreased, but also the proliferation was stimulated by oxidized low-density lipoprotein, which was decreased after downregulation of Hoxc6 expression in VSMCs in rat. Consecutively, the expression of PCNA protein was decreased, while that of p53 was increased. These results indicated that Hoxc6 is probably involved in AS via p53 and PCNA by affecting the proliferation and migration of VSMCs.
Collapse
Affiliation(s)
- Xiangshu Long
- Medical College, Guizhou University, Guiyang 550025, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Ganhua You
- Medical College, Guizhou University, Guiyang 550025, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Qiang Wu
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yu Zhou
- Medical College, Guizhou University, Guiyang 550025, China
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Fuxun Yu
- Department of Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yan Xiao
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Shiyan Deng
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Fang Song
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jing Huang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Maobo Tian
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| |
Collapse
|
14
|
Wang C, Zhao J, Nan X, Guo Z, Huang S, Wang X, Sun F, Ma S. Long noncoding RNA CASC2 inhibits ox-LDL-mediated vascular smooth muscle cells proliferation and migration via the regulation of miR-532-3p/PAPD5. Mol Med 2020; 26:74. [PMID: 32698757 PMCID: PMC7374889 DOI: 10.1186/s10020-020-00200-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/13/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Studies have demonstrated that long noncoding RNAs (lncRNAs) have essential impacts on the development of atherosclerosis (AS). This study aimed to identify the role and functional mechanism of lncRNA CASC2 in the development and migration of vascular smooth muscle cells (VSMCs). METHOD The serum of 40 pairs of AS patients and healthy volunteers were collected and the expression of CASC2 was evaluated. qRT-PCR and western blotting were carried out to examine the expression levels of at mRNA and protein level, repectively. Cell proliferation assay, colony formation assay, transwell migration assay, dual-luciferase reporter assay, and wound healing assay were conducted to evaluate cell proliferation, colony formation, migration, transcription, targeting, and self-restoration. RESULTS The expression levels of CASC2 were decreased, while the expression levels of miR-532-3p were elevated in AS patient samples and VSMCs. Overexpression of CASC2 inhibited the proliferation and migration of VSMCs and enhanced cell apoptosis. CASC2 inhibited the expression of miR-532-3p, and inversely upregulated the expression of PAPD5, which was a target of miR-532-3p. In addition, knockdown of miR-532-3p-mimic and PAPD5 could attenuate the impact of overexpression of CASC2 on proliferation, migration, and apoptosis in ox-LDL-VSMCs. CONCLUSION CASC2 suppressed cell reproduction and promoted cell apoptosis by regulating the miR-532-3p/PAPD5 axis in ox-LDL-mediated VSMCs. This might be important for AS therapeutics.
Collapse
Affiliation(s)
- Chenjing Wang
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China.
| | - Jin Zhao
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| | - Xiaodong Nan
- Department of intensive care unit, Gansu Provincial Corps Hospital of Chinese People's Armed Police Force, Lanzhou City, Gansu Province, 730050, People's Republic of China
| | - Zhong Guo
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| | - Shuangsheng Huang
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| | - Xiaokun Wang
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| | - Feng Sun
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| | - Shijie Ma
- School of Basic Medical Sciences, Northwest Minzu University Health Science Center, No. 1, XibeiXincun Chengguan District, Lanzhou City, Gansu Province, 730030, People's Republic of China
| |
Collapse
|
15
|
MiR-135a-5p inhibits vascular smooth muscle cells proliferation and migration by inactivating FOXO1 and JAK2 signaling pathway. Pathol Res Pract 2020; 224:153091. [PMID: 34174548 DOI: 10.1016/j.prp.2020.153091] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/12/2020] [Accepted: 06/26/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND It is reportedly demonstrated that miR-135a-5p plays a critical role in cancer cells, macrophages, and endothelia cells. However, little is known concerning the function of miR-135a-5p in vascular smooth muscle cells (VSMCs) and atherosclerosis (AS). METHODS Human VSMCs and male C57BL/6 mice were used for establishing AS cell models and animal models. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expressions of miR-135a-5p, forkhead box O1 (FOXO1) mRNA, and Janus kinase 2 (JAK2) mRNA. CCK-8, BrdU, and Transwell assays were used to detect cell migration and proliferation. Cell cycle and apoptosis were analyzed using flow cytometry. The interactions among miR-135a-5p, FOXO1 and JAK2 were validated employing Western blot, qRT-PCR and Luciferase reporter gene assay. RESULTS The expression of miR-135a-5p was significantly decreased in serum samples of AS patients, VSMCs treated with ox-LDL and AS mice models. The overexpression of miR-135a-5p induced VSMCs cycle arrest and apoptosis, and inhibited proliferation and migration. Further experiments confirmed that miR-135a-5p could target and repress FOXO1/CyclinD1 and JAK2/STAT3 pathway. Additionally, the associations among miR-135a-5p, FOXO1/Cyclin D1 and JAK2/STAT3 were validated using animal models. CONCLUSION MiR-135a-5p suppresses VSMCs proliferation and migration induced by ox-LDL via targeting and activating FOXO1/Cyclin D1 and JAK2/STAT3 signaling pathways.
Collapse
|
16
|
Xu SN, Zhou X, Zhu CJ, Qin W, Zhu J, Zhang KL, Li HJ, Xing L, Lian K, Li CX, Sun Z, Wang ZQ, Zhang AJ, Cao HL. Nϵ-Carboxymethyl-Lysine Deteriorates Vascular Calcification in Diabetic Atherosclerosis Induced by Vascular Smooth Muscle Cell-Derived Foam Cells. Front Pharmacol 2020; 11:626. [PMID: 32499695 PMCID: PMC7243476 DOI: 10.3389/fphar.2020.00626] [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: 02/03/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022] Open
Abstract
Nϵ-carboxymethyl-lysine (CML), an advanced glycation end product, is involved in vascular calcification (VC) in diabetic atherosclerosis. This study aimed to investigate the effects of CML on VC in diabetic atherosclerosis induced by vascular smooth muscle cell (VSMC)–derived foam cells. Human studies, animal studies and cell studies were performed. The human study results from 100 patients revealed a poor blood glucose and lipid status and more severe coronary lesions and stenosis in patients with coronary artery disease and diabetes mellitus. Intraperitoneal injection of streptozotocin combined with a high-fat diet was used to build a diabetic atherosclerosis model in ApoE−/− mice. The animal study results indicated that CML accelerated VC progression in diabetic atherosclerosis by accelerating the accumulation of VSMC-derived foam cells in ApoE−/− mice. The cell study results illustrated that CML induced VSMC-derived foam cells apoptosis and aggravated foam cells calcification. Consistent with this finding, calcium content and the expression levels of alkaline phosphatase, bone morphogenetic protein 2 and runt-related transcription factor 2 were significantly elevated in A7r5 cells treated with oxidation-low-density lipoprotein and CML. Thus, we concluded that CML promoted VSMC-derived foam cells calcification to aggravate VC in diabetic atherosclerosis, providing evidence for the contribution of foam cells to diabetic VC.
Collapse
Affiliation(s)
- Sui-Ning Xu
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Xin Zhou
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Cun-Jun Zhu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Qin
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Jie Zhu
- Department of Cardiology, Affiliated Luan Hospital of Anhui Medical University, Luan, China
| | - Ke-Lin Zhang
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Hui-Jin Li
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Lu Xing
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Kun Lian
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Cheng-Xiang Li
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhong-Qun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - An-Ji Zhang
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Hui-Ling Cao
- Department of Cardiology, The First Affiliated Hospital, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| |
Collapse
|
17
|
Correlation between Galectin-3 and Adverse Outcomes in Myocardial Infarction Patients: A Meta-Analysis. Cardiol Res Pract 2020; 2020:7614327. [PMID: 32455000 PMCID: PMC7229552 DOI: 10.1155/2020/7614327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/04/2020] [Accepted: 04/13/2020] [Indexed: 11/18/2022] Open
Abstract
Background Acute myocardial infarction (AMI) is a disease with high morbidity and mortality. Some new biomarkers can help us to improve the life quality and prognosis of AMI patients. Objective We therefore performed a systematic review and meta-analysis on the use of galectin-3 (gal3) for assessing prognosis of AMI patients. Methods We searched Medline, Embase, Web of Science, Cochrane Library, SinoMed, China National Knowledge Infrastructure (CNKI), and Wanfang database up to June 2019. Trials included using galectin-3 to estimate prognosis in myocardial infarction (MI) patients. Results We identified 10 trails with a total of 2809 participants. The negative correlation between galectin-3 and left ventricular ejection fraction (LVEF) was significant in 505 AMI patients (Fisher's Z -0.22, 95% CI: -0.34, -0.09). The correlation between galectin-3 and infarct size was not significant in 119 patients (Fisher's Z 0.12, 95% CI: -0.36, 0.60). Higher galectin-3 was associated with increased all-cause mortality in 2343 AMI patients (Fisher's Z 1.58, 95% CI: 1.23, 2.03). Conclusion The limited evidence suggests that galectin-3 is likely to predict the adverse outcomes in MI patients, but it is not significantly correlated with infarct size after MI. More high-quality trials with longer-term follow-up are still needed to confirm this finding.
Collapse
|
18
|
Lorenzo-Almorós A, Pello A, Aceña Á, Martínez-Milla J, González-Lorenzo Ó, Tarín N, Cristóbal C, Blanco-Colio LM, Martín-Ventura JL, Huelmos A, Gutiérrez-Landaluce C, López-Castillo M, Kallmeyer A, Cánovas E, Alonso J, López Bescós L, Egido J, Lorenzo Ó, Tuñón J. Galectin-3 Is Associated with Cardiovascular Events in Post-Acute Coronary Syndrome Patients with Type-2 Diabetes. J Clin Med 2020; 9:jcm9041105. [PMID: 32294902 PMCID: PMC7230213 DOI: 10.3390/jcm9041105] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction: Type-2 diabetes mellitus (T2DM) is associated with early and severe atherosclerosis. However, few biomarkers can predict cardiovascular events in this population. Methods: We followed 964 patients with coronary artery disease (CAD), assessing plasma levels of galectin-3, monocyte chemoattractant protein-1 (MCP-1), and N-terminal fragment of brain natriuretic peptide (NT-proBNP) at baseline. The secondary outcomes were acute ischemia and heart failure or death. The primary outcome was the combination of the secondary outcomes. Results. Two hundred thirty-two patients had T2DM. Patients with T2DM showed higher MCP-1 (144 (113–195) vs. 133 (105–173) pg/mL, p = 0.006) and galectin-3 (8.3 (6.5–10.5) vs. 7.8 (5.9–9.8) ng/mL, p = 0.049) levels as compared to patients without diabetes. Median follow-up was 5.39 years (2.81–6.92). Galectin-3 levels were associated with increased risk of the primary outcome in T2DM patients (Hazard ratio (HR) 1.57 (1.07–2.30); p = 0.022), along with a history of cerebrovascular events. Treatment with clopidogrel was associated with lower risk. In contrast, NT-proBNP and MCP-1, but not galectin-3, were related to increased risk of the event in nondiabetic patients (HR 1.21 (1.04–1.42); p = 0.017 and HR 1.23 (1.05–1.44); p = 0.012, respectively), along with male sex and age. Galectin-3 was also the only biomarker associated with the development of acute ischemic events and heart failure or death in T2DM patients, while, in nondiabetics, MCP-1 and NT-proBNP, respectively, were related to these events. Conclusion: In CAD patients, galectin-3 plasma levels are associated with cardiovascular events in patients with T2DM, and MCP-1 and NT-proBNP in those without T2DM.
Collapse
Affiliation(s)
- Ana Lorenzo-Almorós
- Department of Internal Medicine, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain;
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
| | - Ana Pello
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Álvaro Aceña
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Juan Martínez-Milla
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Óscar González-Lorenzo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Nieves Tarín
- Department of Cardiology, Hospital Universitario de Móstoles, 28935 Madrid, Spain;
| | - Carmen Cristóbal
- Department of Cardiology, Hospital de Fuenlabrada, 28942 Madrid, Spain; (C.C.); (C.G.-L.)
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
| | - Luis M Blanco-Colio
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
| | - José Luis Martín-Ventura
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
| | - Ana Huelmos
- Department of Cardiology, Hospital Universitario Fundación Alcorcón, 28922 Madrid, Spain;
| | | | - Marta López-Castillo
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
| | - Andrea Kallmeyer
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Ester Cánovas
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
| | - Joaquín Alonso
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
- Department of Cardiology, Hospital de Getafe, 28905 Madrid, Spain
| | - Lorenzo López Bescós
- Department of Medicine, Rey Juan Carlos University, Alcorcón, 28943 Madrid, Spain; (J.A.); (L.L.B.)
| | - Jesús Egido
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de diabetes y enfermedades metabólicas asociadas (CIBERDEM), 28040 Madrid, Spain
| | - Óscar Lorenzo
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de diabetes y enfermedades metabólicas asociadas (CIBERDEM), 28040 Madrid, Spain
| | - Jose Tuñón
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (L.M.B.-C.); (J.L.M.-V.); (J.E.); (Ó.L.)
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.P.); (Á.A.); (J.M.-M.); (Ó.G.-L.); (M.L.-C.); (A.K.); (E.C.)
- Department of Medicine, School of Medicine, Universidad Autónoma, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
19
|
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: 48] [Impact Index Per Article: 12.0] [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
|
20
|
Roles of galectin-3 in metabolic disorders and tumor cell metabolism. Int J Biol Macromol 2020; 142:463-473. [DOI: 10.1016/j.ijbiomac.2019.09.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
|
21
|
He K, Sun H, Zhang J, Zheng R, Gu J, Luo M, Shao Y. Rab7‑mediated autophagy regulates phenotypic transformation and behavior of smooth muscle cells via the Ras/Raf/MEK/ERK signaling pathway in human aortic dissection. Mol Med Rep 2019; 19:3105-3113. [PMID: 30816458 PMCID: PMC6423587 DOI: 10.3892/mmr.2019.9955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023] Open
Abstract
Autophagy regulates the metabolism, survival and function of numerous types of cell, including cells that comprise the cardiovascular system. The dysfunction of autophagy has been demonstrated in atherosclerosis, restenotic lesions and hypertensive vessels. As a member of the Ras GTPase superfamily, Rab7 serves a significant role in the regulation of autophagy. The present study evaluated how Rab7 affects the proliferation and invasion, and phenotypic transformations of aortic dissection (AD) smooth muscle cells (SMCs) via autophagy. Rab7 was overexpressed in AD tissues and the percentage of synthetic human aortic SMCs (HASMCs) was higher in AD tissues compared with NAD tissues. Downregulation of Rab7 decreased cell growth, reduced the number of invasive cells and decreased the percentage cells in the G1 phase. Autophagy of HASMCs was inhibited following Rab7 knockdown. Inhibition of autophagy with 3‑methyladenine or Rab7 knockdown suppressed the phenotypic conversion of contractile to synthetic HASMCs. The action of Rab7 may be mediated by inhibiting the Ras/Raf/mitogen‑activated protein kinase (MAPK) kinase (MEK)/extracellular signal related kinase (ERK) signaling pathway. In conclusion, the results revealed that Rab7‑mediated autophagy regulated the behavior of SMCs and the phenotypic transformations in AD via activation of the Ras/Raf/MEK/ERK signaling pathway. The findings of the present study may improve understanding of the role Rab7 in the molecular etiology of AD and suggests the application of Rab7 as a novel therapeutic target in the treatment of human AD.
Collapse
Affiliation(s)
- Keshuai He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Haoliang Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Junjie Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Rui Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Jiaxi Gu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Ming Luo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
22
|
Tan KCB, Cheung CL, Lee ACH, Lam JKY, Wong Y, Shiu SWM. Galectin-3 and risk of cardiovascular events and all-cause mortality in type 2 diabetes. Diabetes Metab Res Rev 2019; 35:e3093. [PMID: 30378236 DOI: 10.1002/dmrr.3093] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/18/2018] [Accepted: 10/25/2018] [Indexed: 12/23/2022]
Abstract
AIMS Recent clinical studies have shown that galectin-3 is a prognostic indicator in patients with coronary heart disease and in patients with heart failure. Experimental data suggest that galectin-3 may play a role in atherogenesis. We have evaluated whether serum galectin-3 level is associated with cardiovascular outcome in type 2 diabetes. MATERIALS AND METHODS Galectin-3 was measured in baseline samples in 1495 persons with type 2 diabetes. The primary cardiovascular outcome, incident cardiovascular events, was defined as first non-fatal myocardial infarction, non-fatal stroke, coronary revascularization, or death from cardiovascular cause. The secondary outcome was all-cause mortality. RESULTS At baseline, 12% of the subjects had prevalent cardiovascular disease. Serum galectin-3 was increased in the group with incident cardiovascular events compared with those who remained free of events during follow up (9.03 ± 2.98 ng/mL vs 8.15 ± 2.76, P < 0.01). Serum galectin-3 was also significantly increased in those subjects with a fatal outcome. The hazard ratios (HR) for cardiovascular events and all-cause mortality for individuals in the top quartile were 2.50 (95% CI 1.87, 3.36, P < 0.001) and 3.92 (95%CI 2.55, 6.01, P < 0.001), respectively. In a multivariate Cox regression analysis including traditional risk factors, log (eGFR), baseline albuminuria, and cardiovascular disease status, the HR per standard deviation change in galectin-3 was 1.13 (95% CI 1.02, 1.26, P = 0.02) for cardiovascular events and 1.17 (95% CI 1.01, 1.35, P = 0.04) for all-cause mortality. CONCLUSIONS Serum galectin-3 is associated with adverse cardiovascular outcomes in persons with type 2 diabetes independent of traditional risk factors.
Collapse
Affiliation(s)
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong
| | - Alan C H Lee
- Department of Medicine, University of Hong Kong, Hong Kong
| | - Joanne K Y Lam
- Department of Medicine, University of Hong Kong, Hong Kong
| | - Ying Wong
- Department of Medicine, University of Hong Kong, Hong Kong
| | - Sammy W M Shiu
- Department of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
23
|
Chen X, Lin J, Hu T, Ren Z, Li L, Hameed I, Zhang X, Men C, Guo Y, Xu D, Zhan Y. Galectin-3 exacerbates ox-LDL-mediated endothelial injury by inducing inflammation via integrin β1-RhoA-JNK signaling activation. J Cell Physiol 2018; 234:10990-11000. [PMID: 30536538 PMCID: PMC6590151 DOI: 10.1002/jcp.27910] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022]
Abstract
Oxidized low‐density lipoprotein (Ox‐LDL)‐induced endothelial cell injury plays a crucial role in the pathogenesis of atherosclerosis (AS). Plasma galectin‐3 (Gal‐3) is elevated inside and drives diverse systemic inflammatory disorders, including cardiovascular diseases. However, the exact role of Gal‐3 in ox‐LDL‐mediated endothelial injury remains unclear. This study explores the effects of Gal‐3 on ox‐LDL‐induced endothelial dysfunction and the underlying molecular mechanisms. In this study, Gal‐3, integrin β1, and GTP‐RhoA in the blood and plaques of AS patients were examined by ELISA and western blot respectively. Their levels were found to be obviously upregulated compared with non‐AS control group. CCK8 assay and flow cytometry analysis showed that Gal‐3 significantly decreased cell viability and promoted apoptosis in ox‐LDL‐treated human umbilical vascular endothelial cells (HUVECs). The upregulation of integrinβ1, GTP‐RhoA, p‐JNK, p‐p65, p‐IKKα, and p‐IKKβ induced by ox‐LDL was further enhanced by treatment with Gal‐3. Pretreatment with Gal‐3 increased expression of inflammatory factors (interleukin [IL]‐6, IL‐8, and IL‐1β), chemokines(CXCL‐1 and CCL‐2) and adhesion molecules (VCAM‐1 and ICAM‐1). Furthermore, the promotional effects of Gal‐3 on NF‐κB activation and inflammatory factors in ox‐LDL‐treated HUVECs were reversed by the treatments with integrinβ1‐siRNA or the JNK inhibitor. We also found that integrinβ1‐siRNA decreased the protein expression of GTP‐RhoA and p‐JNK, while RhoA inhibitor partially reduced the upregulated expression of p‐JNK induced by Gal‐3. In conclusion, our finding suggests that Gal‐3 exacerbates ox‐LDL‐mediated endothelial injury by inducing inflammation via integrin β1‐RhoA‐JNK signaling activation.
Collapse
Affiliation(s)
- Xiumei Chen
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianzhong Lin
- Department of Urology and Central Laboratory, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Hu
- Department of Cancer Research, The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Zhiyun Ren
- Department of Urology and Central Laboratory, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Linnan Li
- Department of Cancer Research, Academy of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Irbaz Hameed
- Department of Cardiothoracic Surgery, New York Presbyterian Hospital Weill cornell Medicine, New York, New York
| | - Xiaoyu Zhang
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Men
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Guo
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Xu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiyang Zhan
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
SIRT7 Regulates the Vascular Smooth Muscle Cells Proliferation and Migration via Wnt/ β-Catenin Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4769596. [PMID: 30627559 PMCID: PMC6304541 DOI: 10.1155/2018/4769596] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/09/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
Abstract
A huge amount of evidence indicates that sirtuin 7 (SIRT7), a key mediator of many cellular activities, plays a crucial role in the pathogenesis of various diseases. However, little is known about the role of SIRT7 in atherosclerosis. This study investigated the potential role of SIRT7 in regulating the proliferation and migration of human vascular smooth muscle cells (HAVSMCs) and its possible molecular mechanism. In this study, human vascular smooth muscle cells (HAVSMCs) were induced by oxidized low-density lipoprotein (ox-LDL) to establish atherosclerosis (AS) cell model. Immunofluorescence staining and Western blot were used to detect the level of α-SMA expression, which was a marker protein in AS. In addition, RT-qPCR and Western blot assay were applied for exploring the mRNA and protein expression levels of SIRT7, Wnt, β-catenin, and cyclin D1 after knockdown or overexpression of SIRT7. And, furthermore, Cell Counting Kit-8 assay, flow cytometry, and wound-healing assay were used to assess HAVSMCs proliferation, cell cycle, and migration. Dickkopf-1 (DKK-1), a secretory glycoprotein that can block Wnt/β-catenin pathway, was used in SIRT7 overexpression HAVSMCs; subsequently cells proliferation and migration were assessed by Cell Counting Kit-8 assay, flow cytometry analysis, and wound-healing assay. We found that knockdown of SIRT7 significantly promoted cell proliferation and migration, decreased the percentages of cells in the G1 and G2 phases, and increased those in the S phase and downregulated the protein expression levels of Wnt, β-catenin, and cyclin D1, while overexpression of SIRT7 had reverse results. After treatment with Wnt/beta-catenin pathway inhibitor DKK-1 in SIRT7 overexpression HAVSMCs, cell proliferation and migration were increased, respectively. In conclusion, SIRT7 inhibited HAVSMCs proliferation and migration via enhancing Wnt/β-catenin activation, which provided a novel therapeutic strategy for antiatherosclerosis.
Collapse
|
25
|
Suthahar N, Meijers WC, Silljé HH, Ho JE, Liu FT, de Boer RA. Galectin-3 Activation and Inhibition in Heart Failure and Cardiovascular Disease: An Update. Theranostics 2018; 8:593-609. [PMID: 29344292 PMCID: PMC5771079 DOI: 10.7150/thno.22196] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/24/2017] [Indexed: 12/15/2022] Open
Abstract
Galectin-3 is a versatile protein orchestrating several physiological and pathophysiological processes in the human body. In the last decade, considerable interest in galectin-3 has emerged because of its potential role as a biotarget. Galectin-3 is differentially expressed depending on the tissue type, however its expression can be induced under conditions of tissue injury or stress. Galectin-3 overexpression and secretion is associated with several diseases and is extensively studied in the context of fibrosis, heart failure, atherosclerosis and diabetes mellitus. Monomeric (extracellular) galectin-3 usually undergoes further "activation" which significantly broadens the spectrum of biological activity mainly by modifying its carbohydrate-binding properties. Self-interactions of this protein appear to play a crucial role in regulating the extracellular activities of this protein, however there is limited and controversial data on the mechanisms involved. We therefore summarize (recent) literature in this area and describe galectin-3 from a binding perspective providing novel insights into mechanisms by which galectin-3 is known to be "activated" and how such activation may be regulated in pathophysiological scenarios.
Collapse
Affiliation(s)
- Navin Suthahar
- University Medical Center Groningen, University of Groningen, Department of Cardiology, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Wouter C. Meijers
- University Medical Center Groningen, University of Groningen, Department of Cardiology, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Herman H.W. Silljé
- University Medical Center Groningen, University of Groningen, Department of Cardiology, PO Box 30.001, 9700 RB Groningen, the Netherlands
| | - Jennifer E. Ho
- Massachusetts General Hospital, Cardiovascular Research Center, Boston, MA, USA
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Rudolf A. de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, PO Box 30.001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
26
|
Andrejić O, Vučić R, Apostolović S, Pavlović M, Stokanović D, Nikolić V, Jevtović-Stoimenov T, Momčilović S. The Factors Influencing Galectin-3 Levels in Acute Coronary Syndrome with Decreased Left Ventricular Function. ACTA FACULTATIS MEDICAE NAISSENSIS 2017. [DOI: 10.1515/afmnai-2017-0034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Summary
The aim of our study was to determine the factors influencing galectin-3 levels in patients with acute coronary syndrome and decreased left ventricular ejection fraction. We collected material from 37 successive patients with acute coronary syndrome and decreased left ventricular ejection fraction, of which 19 patients had atrial fibrillation, and 18 patients who were without atrial fibrillation constituted a control group. Blood samples used for the biochemical measurements were obtained on the third day from acute coronary syndrome. We used Statistical Package for Social Sciences for data analysis. A p-value less than 0.05 was considered to be a measure of statistical significance. Galectin-3 concentration is directly correlated with age and B-type natriuretic peptide level. Also, our results showed an inverse correlation between galectin-3 and total body weight, body mass index, body surface area and creatinine clearance. The following variables were found to be significant predictors of galectin-3 level: decreased left ventricular ejection fraction, total body weight, LDL concentration and body mass index. We identified factors that can predict a decrease in the left ventricular ejection fraction below 45% after acute coronary syndrome: atrial fibrillation increases the risk by almost six times, and urea concentration increases the risk by 1.2 times for each unit. Left ventricular ejection fraction below 45%, TBW, body mass index and LDL level are good predictors of galectin-3 concentration in patients with ACS and decreased left ventricular ejection fraction. Atrial fibrillation could be a predictive marker of decreased left ventricular ejection fraction.
Collapse
|
27
|
Galectin-3 in acute coronary syndrome. Clin Biochem 2017; 50:797-803. [DOI: 10.1016/j.clinbiochem.2017.04.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/20/2017] [Accepted: 04/25/2017] [Indexed: 11/18/2022]
|
28
|
Activation of CD137 signaling accelerates vascular calcification in vivo and vitro. Int J Cardiol 2017; 230:198-203. [DOI: 10.1016/j.ijcard.2016.12.174] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/18/2016] [Accepted: 12/25/2016] [Indexed: 01/08/2023]
|
29
|
Saccon F, Gatto M, Ghirardello A, Iaccarino L, Punzi L, Doria A. Role of galectin-3 in autoimmune and non-autoimmune nephropathies. Autoimmun Rev 2016; 16:34-47. [PMID: 27666815 DOI: 10.1016/j.autrev.2016.09.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
Galectins are evolutionary conserved β-galactoside binding proteins with a carbohydrate-recognition domain (CRD) of approximately 130 amino acids. In mammals, 15 members of the galectin family have been identified and classified into three subtypes according to CRD organization: prototype, tandem repeat-type and chimera-type galectins. Galectin-3 (gal-3) is the only chimera type galectin in vertebrates containing one CRD linked to an unusual long N-terminal domain which displays non-lectin dependent activities. Although recent studies revealed unique, pleiotropic and context-dependent functions of gal-3 in both extracellular and intracellular space, gal-3 specific pathways and its ligands have not been clearly defined yet. In the kidney gal-3 is involved in later stages of nephrogenesis as well as in renal cell cancer. However, gal-3 has recently been associated with lupus glomerulonephritis, with Familial Mediterranean Fever-induced proteinuria and renal amyloidosis. Gal-3 has been studied in experimental acute kidney damage and in the subsequent regeneration phase as well as in several models of chronic kidney disease, including nephropathies induced by aging, ischemia, hypertension, diabetes, hyperlipidemia, unilateral ureteral obstruction and chronic allograft injury. Because of the pivotal role of gal-3 in the modulation of immune system, wound repair, fibrosis and tumorigenesis, it is not surprising that gal-3 can be an intriguing prognostic biomarker as well as a promising therapeutic target in a great variety of diseases, including chronic kidney disease, chronic heart failure and cardio-renal syndrome. This review summarizes the functions of gal-3 in kidney pathophysiology focusing on the reported role of gal-3 in autoimmune diseases.
Collapse
Affiliation(s)
- Francesca Saccon
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy
| | - Mariele Gatto
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy
| | - Anna Ghirardello
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy
| | - Luca Iaccarino
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy
| | - Leonardo Punzi
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy
| | - Andrea Doria
- Division of Rheumatology, Department of Medicine (DIMED), University of Padova, Italy.
| |
Collapse
|