1
|
Mu Y, Wei M, Liu Y, Fan H, Yuan J, Cai S, He H, Gou J, Tang X, Yin T, Zhang Y. Lactoferrin-functionalized PEGylation liposomes loaded with norcantharidin acid for targeted therapy of hepatocellular carcinoma. Int J Pharm 2025; 671:125245. [PMID: 39842742 DOI: 10.1016/j.ijpharm.2025.125245] [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: 09/08/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Norcantharidin (NCTD), an antitumor agent with an increased leukocyte function, has been used for the treatment of hepatocellular carcinoma (HCC) in clinical. However, the clinical application of NCTD is limited due to its inadequate hydrophilicity and lipophilicity, short half-life (t1/2), as well as adverse effects such as vascular irritation, cardiotoxicity, and nephrotoxicity. Herein, a lactoferrin (Lf) and DSPE-mPEG2000 functionalized liposomes loaded with norcantharidic acid (NCA), an active metabolite of NCTD, was constructed for the targeted therapy of HCC. In this study, blank PEGylated liposomes were prepared using the film hydration method, and the NCA was loaded by calcium acetate active loading method to increase the encapsulation efficiency (EE). Subsequently, lactoferrin was covalently coupled to DSPE-PEG2000-COOH activated by EDC and NHS. In addition, the in vivo pharmacokinetics and pharmacodynamics were investigated in Sprague-Dawley (SD) rats and H22 tumor-bearing BALB/c mice, respectively. As expected, the encapsulation efficiency measurement showed that the encapsulation efficiency of the NCA liposomes was 89.3±1.25 %, and the coupling efficiency of lactoferrin was more than 65.97 %. Additionally, the variations in both the dynamic size and encapsulation efficiency of norcantharidic acid liposomes in long-term storage stability and serum stability studies did not exceed 10 %. Furthermore, the pharmacokinetics and pharmacodynamics results showed that, the NCA-Lips-Lf were able to significantly improve antitumor activity by enhancing tumor-targeting accumulation and prolonging circulation time in the body compared to the sodium demethylcantharidate for injection (Na2DCA). Notably, the AUC0-48 and the t1/2 of NCA-Lips-Lf increased 4.28-time and 5.17-time in comparison to those of NCA-sol, respectively. The tumor inhibition rate of NCA-Lips-Lf (85.29 %) was significantly higher than that of sodium demethylcantharidate for injection (Na2DCA) (59.13 %), without obvious vascular irritation, cardiotoxicity and nephrotoxicity. In conclusion, NCA-Lips-Lf have the potential to eliminate hepatocellular carcinoma more effectively with fewer side effects than Na2DCA, which further advances the clinical application of norcantharidin-related drugs.
Collapse
Affiliation(s)
- Yinling Mu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Mingli Wei
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Yuxin Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Hongxia Fan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Jingjing Yuan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shunqiao Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Haibing He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Jingxin Gou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| | - Tian Yin
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Yu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110116, China
| |
Collapse
|
2
|
Peng Z, Luo XY, Li X, Li Y, Wu Y, Tian Y, Pan B, Petrovic A, Kosanovic D, Schermuly RT, Ruppert C, Günther A, Zhang Z, Qiu C, Li Y, Pu J, Li X, Chen AF. Cathepsin L Promotes Pulmonary Hypertension via BMPR2/GSDME-Mediated Pyroptosis. Hypertension 2024; 81:2430-2443. [PMID: 39403807 DOI: 10.1161/hypertensionaha.124.22903] [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: 02/18/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a fatal progressive disease characterized by pulmonary endothelial injury and occlusive pulmonary vascular remodeling. Lysosomal protease cathepsin L degrades essential molecules to participate in the human pathophysiological process. BMPR2 (bone morphogenetic protein type II receptor) deficiency, an important cause of PH, results from mutational inactivation or excessive lysosomal degradation and induces caspase-3-mediated cell death. Given recent evidence that pyroptosis, as a new form of programmed cell death, is induced by caspase-3-dependent GSDME (gasdermin E) cleavage, we hypothesized that cathepsin L might promote PH through BMPR2/caspase-3/GSDME axis-mediated pyroptosis. METHODS Cathepsin L expression was evaluated in the lungs and plasma of patients with pulmonary arterial hypertension. The role of cathepsin L in the progression of PH and vascular remodeling was assessed in vivo. Small interfering RNA, specific inhibitors, and lentiviruses were used to explore the mechanisms of cathepsin L on human pulmonary arterial endothelial cell dysfunction. RESULTS Cathepsin L expression is elevated in pulmonary artery endothelium from patients with idiopathic pulmonary arterial hypertension and experimental PH models. Genetic ablation of cathepsin L in PH rats relieved right ventricular systolic pressure, pulmonary vascular remodeling, and right ventricular hypertrophy, also restoring endothelial integrity. Mechanistically, cathepsin L promotes caspase-3/GSDME-mediated endothelial cell pyroptosis and represses BMPR2 signaling activity. Cathepsin L degrades BMPR2 via the lysosomal pathway, and restoring BMPR2 signaling prevents the pro-pyroptotic role of cathepsin L in PAECs and experimental PH models. CONCLUSIONS These results show for the first time that cathepsin L promotes the development of PH by degrading BMPR2 to induce caspase-3/GSDME-mediated endothelial pyroptosis.
Collapse
Affiliation(s)
- Zhouyangfan Peng
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Xue-Yang Luo
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Xinyi Li
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Yapei Li
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Health Management Medicine Center, the Third Xiangya Hospital (Yapei Li., Ying Li.), Central South University, Changsha, China
| | - Yusi Wu
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Yuyang Tian
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bingjie Pan
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Aleksandar Petrovic
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Djuro Kosanovic
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University, Russia (D.K.)
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Andreas Günther
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Zhen Zhang
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
| | - Chengfeng Qiu
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Ying Li
- Health Management Medicine Center, the Third Xiangya Hospital (Yapei Li., Ying Li.), Central South University, Changsha, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (J.P.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaohui Li
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Alex F Chen
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
3
|
Zhang W, Xiang C, Liu B, Hou F, Zheng Z, Chen Z, Suo L, Feng G, Gu J. The value of systemic immune inflammation index, white blood cell to platelet ratio, and homocysteine in predicting the instability of small saccular intracranial aneurysms. Sci Rep 2024; 14:24312. [PMID: 39414876 PMCID: PMC11484959 DOI: 10.1038/s41598-024-74870-y] [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: 05/11/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
Inflammation has a destructive effect on the homeostasis of the vascular wall, which is involved in the formation, growth, and rupture of human intracranial aneurysms (IAs) disease progression. However, inflammation-related markers have not been well studied in the risk stratification of unruptured IAs. The purpose of this study was to investigate the predictive value of serum inflammatory markers in the unstable progression of small saccular intracranial aneurysms (SIAs). This study retrospectively included 275 patients with small SIAs (aneurysm diameter less than or equal to 7 mm), to compare the level difference of serum inflammatory complex marker systemic immune-inflammatory index (SII), white blood cell to platelet ratio (WPR), and homocysteine (Hcy) in patients with stable (asymptomatic unruptured) and unstable (symptomatic unruptured, ruptured) small SIAs. 187 patients (68%) had aneurysm-related compression symptoms and rupture outcomes. In the multivariate logistic regression after adjusting for baseline differences, SII, WPR, and Hcy were independent risk factors for the instability of small SIAs, the prediction model combined with other risk factors (previous stroke history, aneurysm irregularity) showed good predictive ability for the instability of small SIAs, with an area under the curve of 0.905. In addition, correlation analysis showed that SII, WPR, and Hcy also had significant differences in patients with symptomatic unruptured and ruptured small SIAs, and higher inflammation levels often promoted the disease progression of small SIAs. Higher levels of SII, WPR and Hcy can be used as independent predictors of instability of small SIAs. As an economical and convenient biomarker, it is crucial for clinical treatment strategies of stable small SIAs.
Collapse
Affiliation(s)
- Wanwan Zhang
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China
- Department of Clinical Medicine, Henan University, Kaifeng, Henan, People's Republic of China
| | - Chao Xiang
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Boliang Liu
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Fandi Hou
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhanqiang Zheng
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhongcan Chen
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Lina Suo
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Guang Feng
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China.
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Jianjun Gu
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Henan University, Zhengzhou, Henan, People's Republic of China.
- Department of Neurosurgery, Henan Provincial People's Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
4
|
Folate and Vitamin B 12 Deficiency Exacerbate Inflammation during Mycobacterium avium paratuberculosis (MAP) Infection. Nutrients 2023; 15:nu15020261. [PMID: 36678131 PMCID: PMC9865721 DOI: 10.3390/nu15020261] [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: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Folate and vitamin B12 deficiency is highly prevalent among Crohn's disease (CD) patients. Furthermore, CD pathology can be mediated by Mycobacterium avium subsp. paratuberculosis (MAP) infection. However, the direct effect of folate (B9) and cobalamin (B12) deficiency during MAP infection remains uncharacterized. This study investigates how folate and B12 deficiency impedes macrophage apoptosis and exacerbates the inflammation in macrophages infected with MAP isolated from CD patients. Accordingly, we measured folate and B12 in ex vivo plasma samples collected from CD patients with or without MAP infection (N = 35 per group). We also measured the expression of the pro-inflammatory cytokines IL-1β and TNF-α, cellular apoptosis and viability markers, and bacterial viability in MAP-infected macrophages cultured in folate and B12 deficient media. We determined that MAP-positive CD patients have significantly lower plasma folate and B12 in comparison to MAP-negative CD patients [414.48 ± 94.60 pg/mL vs. 512.86 ± 129.12 pg/mL, respectively]. We further show that pro-inflammatory cytokines IL-1β and TNF-α are significantly upregulated during folate and vitamin B12 deprivation following MAP infection by several folds, while supplementation significantly reduces their expression by several folds. Additionally, depletion of folate, B12, and folate/B12 following MAP infection, led to decreased macrophage apoptosis from 1.83 ± 0.40-fold to 1.04 ± 0.08, 0.64 ± 0.12, and 0.45 ± 0.07 in folate-low, B12-low, and folate/B12-low cells, respectively. By contrast, folate and folate/B12 supplementation resulted in 3.38 ± 0.70 and 2.58 ± 0.14-fold increases in infected macrophages. Interestingly, changes in overall macrophage viability were only observed in folate-high, folate/B12-high, and folate/B12-low media, with 0.80 ± 0.05, 0.82 ± 0.02, and 0.91 ± 0.04-fold changes, respectively. Incubation of Caco-2 intestinal epithelial monolayers with supernatant from infected macrophages revealed that folate/B12 deficiency led to increased LDH release independent of oxidative stress. Overall, our results indicate that folate and B12 are key vitamins affecting cell survival and inflammation during MAP infection.
Collapse
|
5
|
Effects of four weeks lasting aerobic physical activity on cardiovascular biomarkers, oxidative stress and histomorphometric changes of heart and aorta in rats with experimentally induced hyperhomocysteinemia. Mol Cell Biochem 2023; 478:161-172. [PMID: 35759142 DOI: 10.1007/s11010-022-04503-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/10/2022] [Indexed: 01/17/2023]
Abstract
The aim of this study was to examine the effects of hyperhomocysteinemia and aerobic physical activity on changes of cardiovascular biomarkers in sera, oxidative stress in cardiac tissue, and histomorphometric parameters of heart and aorta in rats. Experiments were conducted on male Wistar albino rats organized into four groups (n = 10, per group): C (control group): 0.9% NaCl 0.2 mL/day; H (homocysteine group): homocysteine 0.45 µmol/g b.w./day; CPA (control + physical activity group): 0.9% NaCl 0.2 mL/day and a program of physical activity on a treadmill; and HPA (homocysteine + physical activity group) homocysteine 0.45 µmol/g b.w./day and a program of physical activity on a treadmill. Substances were applied subcutaneously twice a day. Lipid peroxidation and relative activity of Mn-superoxide dismutase isoform were significantly higher in active hyperhomocysteinemic rats in comparison to sedentary animals. Atherosclerotic plaques were detected in aorta samples of active hyperhomocysteinemic rats and also, they had increased left ventricle wall and interventricular septum, and transverse diameter of cardiomyocytes compared to sedentary groups. Aerobic physical activity in the condition of hyperhomocysteinemia can lead to increased oxidative stress in cardiac tissue and changes in histomorphometric parameters of the heart and aorta, as well increased lipid parameters and cardiac damage biomarkers in sera of rats.
Collapse
|
6
|
Lecaille F, Chazeirat T, Saidi A, Lalmanach G. Cathepsin V: Molecular characteristics and significance in health and disease. Mol Aspects Med 2022; 88:101086. [PMID: 35305807 DOI: 10.1016/j.mam.2022.101086] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 12/31/2022]
Abstract
Human cysteine cathepsins form a family of eleven proteases (B, C, F, H, K, L, O, S, V, W, X/Z) that play important roles in a considerable number of biological and pathophysiological processes. Among them, cathepsin V, also known as cathepsin L2, is a lysosomal enzyme, which is mainly expressed in cornea, thymus, heart, brain, and skin. Cathepsin V is a multifunctional endopeptidase that is involved in both the release of antigenic peptides and the maturation of MHC class II molecules and participates in the turnover of elastin fibrils as well in the cleavage of intra- and extra-cellular substrates. Moreover, there is increasing evidence that cathepsin V may contribute to the progression of diverse diseases, due to the dysregulation of its expression and/or its activity. For instance, increased expression of cathepsin V is closely correlated with malignancies (breast cancer, squamous cell carcinoma, or colorectal cancer) as well vascular disorders (atherosclerosis, aortic aneurysm, hypertension) being the most prominent examples. This review aims to shed light on current knowledge on molecular aspects of cathepsin V (genomic organization, protein structure, substrate specificity), its regulation by protein and non-protein inhibitors as well to summarize its expression (tissue and cellular distribution). Then the core biological and pathophysiological roles of cathepsin V will be depicted, raising the question of its interest as a valuable target that can open up pioneering therapeutic avenues.
Collapse
Affiliation(s)
- Fabien Lecaille
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| | - Thibault Chazeirat
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Ahlame Saidi
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France
| | - Gilles Lalmanach
- Université de Tours, Tours, France; INSERM, UMR 1100, Centre d'Etude des Pathologies Respiratoires (CEPR), Team "Mécanismes protéolytiques dans l'inflammation", Tours, France.
| |
Collapse
|
7
|
Effects of Personalized Nursing plus Dietary Nursing Management on LP-PLA2, Hcy Levels, and Quality of Life in Elderly Patients with Acute Coronary Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8770996. [PMID: 36034968 PMCID: PMC9402311 DOI: 10.1155/2022/8770996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/03/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022]
Abstract
Objective To explore the effect of individual nursing combined with dietary management on blood glucose levels and quality of life in elderly patients with acute coronary syndrome with diabetes. Method This study included 68 elderly patients who underwent acute coronary syndrome at Department of Emergency in our hospital from January 2018 to January 2020. And they were assigned to a control group (n = 34) treated with individual nursing and a research group (n = 34) treated with individual nursing and dietary management according to the random number table. A comparison of the changes in blood glucose levels, anxiety and depression, quality of life, and nursing satisfaction was made between groups before and after nursing. Result Before nursing, the differences in blood glucose levels, SAS scores, and SDS scores between groups were not considered statistically significant (P > 0.05). After nursing, the blood glucose levels, SAS, and SDS levels of patients in both groups significantly decreased, and the research group presented a higher decrease when compared with the control group (P < 0.05). The short form health survey (SF-36) showed that the research group had higher scores in physical function (PF), general health (GH), vitality (VT), mental health (MH), social function (SF), role-physical (RP), bodily pain (BP), and role-emotional (RE) compared with the control group (P < 0.05). The satisfaction survey presented that the research group had a significantly higher total score than the control group [(91.40 ± 5.23) vs. (86.61 ± 7.14), P < 0.05]. Conclusion The combination of individual nursing and dietary management not only effectively reduces glycosylated hemoglobin levels and anxiety and depression but also wins better nursing satisfaction in the treatment of acute coronary syndrome in elderly patients. Moreover, their quality of life has been significantly improved after discharge.
Collapse
|
8
|
Kim DB, Unenkhuu B, Kim GJ, Kim SW, Kim HS. Cynarin attenuates LPS-induced endothelial inflammation via upregulation of the negative regulator MKP-3. Anim Cells Syst (Seoul) 2022; 26:119-128. [PMID: 35784390 PMCID: PMC9246029 DOI: 10.1080/19768354.2022.2077438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Clinical observations have revealed that non-resolving low-grade inflammation is linked to the pathogenesis of chronic inflammatory diseases, for example arthritis, atherosclerosis, Alzheimer’s disease, diabetes, and chronic kidney disease. Interestingly, low levels of circulating lipopolysaccharides (LPS) derived from the outer membrane of gram-negative bacteria appear to be one of the primary causes of persistent low-grade inflammation. The inner surface of the blood vessels is lined with endothelial cells; therefore, even low levels of circulating LPS can directly activate these cells and elicit specific cellular responses, such as an increase in the expression levels of cell adhesion molecules and proinflammatory mediators. In endothelial cells, LPS exposure results in an inflammatory response through activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinases. Cynarin, a phytochemical found in artichokes, has several pharmacological properties against endothelial inflammation. In the present study, we discovered that cynarin suppressed the LPS-induced increase in the expression levels of vascular cell adhesion molecule-1 and proinflammatory mediators such as monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), and interleukin-1β in EA.hy926 cells. Further, cynarin inhibited the activation of p38 and NF-κB pathways by inducing the negative regulator mitogen-activated protein kinase phosphatase 3 (MKP-3) in LPS-stimulated EA.hy926 cells. In conclusion, cynarin alleviates inflammation by upregulating MKP-3, a negative regulator of p38 and NF-κB, and it may be a therapeutic option for treating endothelial inflammation-related diseases.
Collapse
Affiliation(s)
- Da Bin Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
- Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Banzragchgarav Unenkhuu
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Grace Jisoo Kim
- Yongsan International School of Seoul, Seoul, Republic of Korea
| | - Seung-Woo Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
9
|
Zinellu A, Mangoni AA. Effect of statin treatment on homocysteine concentrations: an updated systematic review and meta-analysis with meta-regression. Expert Rev Clin Pharmacol 2022; 15:443-459. [PMID: 35482022 DOI: 10.1080/17512433.2022.2072293] [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] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Statins might exert atheroprotective effects through lowering the pro-atherogenic amino acid homocysteine. We conducted an updated systematic review and meta-analysis of the effect of statins on circulating homocysteine. METHODS A systematic literature search was conducted in PubMed, Web of Science, and Scopus, from inception to July 2021. The risk of bias was assessed using the Joanna Briggs Institute Critical Appraisal Checklist for analytical studies. Certainty of evidence was assessed using GRADE. RESULTS In 61 treatment arms in 2,218 patients (mean age 55 years, 52% males), statins significantly reduced homocysteine concentrations (weighted mean difference, WMD = -2.46 µmol/L, 95% CI -3.17 to -1.75 µmol/L, p < 0.001; high certainty of evidence). Similar results were observed in a subgroup of 10 randomized placebo-controlled studies (WMD = -2.45 µmol/L, 95% CI -4.43 to -0.47 µmol/L, p = 0.015). The extreme heterogeneity observed was virtually removed in a subgroup of 10 studies using fluorescence polarization immunoassay for homocysteine measurement. There was no publication bias. In sensitivity analysis, the pooled WMD values were not modified when individual studies were sequentially removed. In meta-regression, the WMD was significantly associated with proportion of males and publication year. CONCLUSIONS Statins significantly lower homocysteine concentrations.
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
10
|
Manzine PR, Vatanabe IP, Grigoli MM, Pedroso RV, de Almeida MPOMEP, de Oliveira DDSMS, Crispim Nascimento CM, Peron R, de Souza Orlandi F, Cominetti MR. Potential Protein Blood-Based Biomarkers in Different Types of Dementia: A Therapeutic Overview. Curr Pharm Des 2022; 28:1170-1186. [DOI: 10.2174/1381612828666220408124809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Biomarkers capable of identifying and distinguishing types of dementia such as Alzheimer's disease (AD), Parkinson's disease dementia (PDD), Lewy body dementia (LBD), and frontotemporal dementia (FTD) have been become increasingly relentless. Studies of possible biomarker proteins in the blood that can help formulate new diagnostic proposals and therapeutic visions of different types of dementia are needed. However, due to several limitations of these biomarkers, especially in discerning dementia, their clinical applications are still undetermined. Thus, the updating of biomarker blood proteins that can help in the diagnosis and discrimination of these main dementia conditions is essential to enable new pharmacological and clinical management strategies, with specificities for each type of dementia. To review the literature concerning protein blood-based AD and non-AD biomarkers as new pharmacological targets and/or therapeutic strategies. Recent findings for protein-based AD, PDD, LBD, and FTD biomarkers are focused on in this review. Protein biomarkers were classified according to the pathophysiology of the dementia types. The diagnosis and distinction of dementia through protein biomarkers is still a challenge. The lack of exclusive biomarkers for each type of dementia highlights the need for further studies in this field. Only after this, blood biomarkers may have a valid use in clinical practice as they are promising to help in diagnosis and in the differentiation of diseases.
Collapse
Affiliation(s)
- Patricia Regina Manzine
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Izabela Pereira Vatanabe
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Marina Mantellatto Grigoli
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Renata Valle Pedroso
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | | | | | | | - Rafaela Peron
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Fabiana de Souza Orlandi
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| | - Márcia Regina Cominetti
- Department of Gerontology, Federal University of Sao Carlos, Brazil. Highway Washington Luis, Km 235. Monjolinho
| |
Collapse
|
11
|
Moretti R, Caruso P. Small Vessel Disease: Ancient Description, Novel Biomarkers. Int J Mol Sci 2022; 23:3508. [PMID: 35408867 PMCID: PMC8998274 DOI: 10.3390/ijms23073508] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Small vessel disease (SVD) is one of the most frequent pathological conditions which lead to dementia. Biochemical and neuroimaging might help correctly identify the clinical diagnosis of this relevant brain disease. The microvascular alterations which underlie SVD have common origins, similar cognitive outcomes, and common vascular risk factors. Nevertheless, the arteriolosclerosis process, which underlines SVD development, is based on different mechanisms, not all completely understood, which start from a chronic hypoperfusion state and pass through a chronic brain inflammatory condition, inducing a significant endothelium activation and a consequent tissue remodeling action. In a recent review, we focused on the pathophysiology of SVD, which is complex, involving genetic conditions and different co-morbidities (i.e., diabetes, chronic hypoxia condition, and obesity). Currently, many points still remain unclear and discordant. In this paper, we wanted to focus on new biomarkers, which can be the expression of the endothelial dysfunction, or of the oxidative damage, which could be employed as markers of disease progression or for future targets of therapies. Therefore, we described the altered response to the endothelium-derived nitric oxide-vasodilators (ENOV), prostacyclin, C-reactive proteins, and endothelium-derived hyperpolarizing factors (EDHF). At the same time, due to the concomitant endothelial activation and chronic neuroinflammatory status, we described hypoxia-endothelial-related markers, such as HIF 1 alpha, VEGFR2, and neuroglobin, and MMPs. We also described blood-brain barrier disruption biomarkers and imaging techniques, which can also describe perivascular spaces enlargement and dysfunction. More studies should be necessary, in order to implement these results and give them a clinical benefit.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy;
| | | |
Collapse
|
12
|
Mitrović A, Senjor E, Jukić M, Bolčina L, Prunk M, Proj M, Nanut MP, Gobec S, Kos J. New inhibitors of cathepsin V impair tumor cell proliferation and elastin degradation and increase immune cell cytotoxicity. Comput Struct Biotechnol J 2022; 20:4667-4687. [PMID: 36147668 PMCID: PMC9459403 DOI: 10.1016/j.csbj.2022.08.046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Identification of novel potent inhibitors of lysosomal cysteine peptidase cathepsin V. New inhibitors of cathepsin V demonstrated antitumor activity. They impair tumor cell proliferation and elastase degradation and increase immune cell cytotoxicity. Cathepsin V inhibitor impaired conversion of immunosuppressive factor cystatin F to its active monomeric form.
Cathepsin V is a human lysosomal cysteine peptidase with specific functions during pathological processes and is as such a promising therapeutic target. Peptidase inhibitors represent powerful pharmacological tools for regulating excessive proteolytic activity in various diseases. Cathepsin V is highly related to cathepsin L but differs in tissue distribution, binding site morphology, substrate specificity, and function. To validate its therapeutic potential and extend the number of potent and selective cathepsin V inhibitors, we used virtual high-throughput screening of commercially available compound libraries followed by an evaluation of kinetic properties to identify novel potent and selective cathepsin V inhibitors. We identified the ureido methylpiperidine carboxylate derivative, compound 7, as a reversible, selective, and potent inhibitor of cathepsin V. It also exhibited the most preferable characteristics for further evaluation with in vitro functional assays that simulate the processes in which cathepsin V is known to play an important role. Compound 7 exerted significant effects on cell proliferation, elastin degradation, and immune cell cytotoxicity. The latter was increased because compound 7 impaired conversion of immunosuppressive factor cystatin F to its active monomeric form. Taken together, our results present novel potent inhibitors of cathepsin V and provide new hit compounds for detailed development and optimization. Further, we demonstrate that cathepsin V is a potential target for new approaches to cancer therapy.
Collapse
Affiliation(s)
- Ana Mitrović
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
- Corresponding author at: Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.
| | - Emanuela Senjor
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Marko Jukić
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Lara Bolčina
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Mateja Prunk
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Matic Proj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Milica Perišić Nanut
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
13
|
Vaccaro JA, Naser SA. The Role of Methyl Donors of the Methionine Cycle in Gastrointestinal Infection and Inflammation. Healthcare (Basel) 2021; 10:healthcare10010061. [PMID: 35052225 PMCID: PMC8775811 DOI: 10.3390/healthcare10010061] [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: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/01/2022] Open
Abstract
Vitamin deficiency is well known to contribute to disease development in both humans and other animals. Nonetheless, truly understanding the role of vitamins in human biology requires more than identifying their deficiencies. Discerning the mechanisms by which vitamins participate in health is necessary to assess risk factors, diagnostics, and treatment options for deficiency in a clinical setting. For researchers, the absence of a vitamin may be used as a tool to understand the importance of the metabolic pathways in which it participates. This review aims to explore the current understanding of the complex relationship between the methyl donating vitamins folate and cobalamin (B12), the universal methyl donor S-adenosyl-L-methionine (SAM), and inflammatory processes in human disease. First, it outlines the process of single-carbon metabolism in the generation of first methionine and subsequently SAM. Following this, established relationships between folate, B12, and SAM in varying bodily tissues are discussed, with special attention given to their effects on gut inflammation.
Collapse
|
14
|
Li W, Pan J, Wei M, Lv Z, Chen S, Qin Y, Li N. Nonocular Influencing Factors for Primary Glaucoma: An Umbrella Review of Meta-Analysis. Ophthalmic Res 2021; 64:938-950. [PMID: 34517373 DOI: 10.1159/000519247] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 08/22/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Glaucoma is the main cause of irreversible blindness worldwide. Still, little is known about nonocular risk factors. We use an umbrella review to examine the meta-analytic evidence of the correlation between nonocular factors and glaucoma. METHOD We searched PubMed and Embase databases up to July 24, 2020. Eligible meta-analyses (MAs) included cohort, case-control, and randomized controlled study designs. Two authors independently extracted the data and evaluated the methodological quality of the MAs. AMSTAR 2 was used to assess the methodological quality of each included MA. RESULTS This umbrella review contains 22 MAs with 22 unique nonocular factors in total. We identified 11 factors that increase the risk of glaucoma: hyperlipidemia, nocturnal dip in blood pressure, infection with Helicobacter pylori, myopia, obstructive sleep apnea syndrome, corneal properties, diabetes, hypertension, hypothyroidism, migraine, and plasma homocysteine. We identified 3 factors that reduce the risk of glaucoma: dietary intake of vitamin A, dietary intake of vitamin C, and short-term statin use. We identified 8 factors that had no association with glaucoma: dietary intake of vitamin B, dietary intake of vitamin E, cigarette smoking, Alzheimer's disease, serum folic acid, serum vitamin B6, serum vitamin B12, and serum vitamin D. CONCLUSIONS In this umbrella review of MAs, evidence was found for associations of various nonocular factors with glaucoma to different degrees. However, risk factors were only mildly associated, suggesting low impact of systemic risk factors. Additional higher quality studies are needed to provide robust evidence.
Collapse
Affiliation(s)
- Wenman Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China,
| | - Jiaxing Pan
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Maoling Wei
- The Center for Evidence-Based Medicine and Clinical Epidemiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiqing Lv
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Sijie Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Qin
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Unenkhuu B, Kim DB, Kim HS. MKP-3 suppresses LPS-induced inflammatory responses in HUVECs via inhibition of p38 MAPK/NF-κB pathway. Anim Cells Syst (Seoul) 2021; 25:235-244. [PMID: 34408812 PMCID: PMC8366647 DOI: 10.1080/19768354.2021.1954551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell dysfunction and inflammatory responses play critical roles in the development of atherosclerosis. Recent data on the processes underlying atherogenesis indicate the substantial role of endotoxins (lipopolysaccharides; LPS) of the intestinal microflora in the initiation and progression of atherosclerosis. Mitogen-activated protein (MAP) kinase phosphatase-3 (MKP-3) is a cytoplasmic dual-specificity protein phosphatase that specifically binds to and inactivates MAP kinases in mammalian cells, but its biological function in endothelial cell dysfunction and inflammatory responses remains largely unknown. The aim of the present study was to investigate the role of MKP-3 in endotoxin-induced endothelial inflammation by western blotting, quantitative polymerase chain reaction, and immunofluorescence. The results of our study demonstrated that MKP-3 overexpression markedly inhibited the adhesion of human monocytic THP-1 cells to human umbilical vein endothelial cells (HUVECs) by downregulating the expression of vascular cell adhesion protein 1 (VCAM-1) and pro-inflammatory cytokines. In contrast, MKP-3-encoding gene knockdown by small interfering RNA (siRNA) exacerbated LPS-induced endothelial dysfunction. Additionally, we found that MKP-3 overexpression inhibited LPS-induced p38 MAPK phosphorylation and decreased the nuclear translocation of nuclear factor kappa B (NF-κB) after LPS treatment, suggesting its implication in the LPS/Toll-like receptor 4 (TLR4)/p38/NF-κB pathway. Overall, these observations suggest that MKP-3 plays a protective role in endothelial dysfunction and may be a therapeutic target.
Collapse
Affiliation(s)
- Banzragchgarav Unenkhuu
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Da Bin Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea.,Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, Republic of Korea
| |
Collapse
|
16
|
Li X, Gao M, Zhu S, Yin L, Zhang B, Qi Y, Zhao Y, Yu Y, Xu L. Hengshun Aromatic Vinegar Ameliorates Vascular Endothelial Injury via Regulating PKCζ-Mediated Oxidative Stress and Apoptosis. Front Nutr 2021; 8:635232. [PMID: 34124116 PMCID: PMC8193096 DOI: 10.3389/fnut.2021.635232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/23/2021] [Indexed: 01/14/2023] Open
Abstract
Vascular endothelial injury (VEI) is an early event of atherosclerosis, and reversing endothelial dysfunction has become a new trend in the prevention and treatment of cardiovascular diseases. Hengshun aromatic vinegar (HSAV), a traditional vinegar, has been reported to have many pharmacological activities, but its effect against VEI and the molecular mechanism are still unknown. In this study, effects of HSAV on VEI were evaluated in H2O2-induced human umbilical vein endothelial cells (HUVECs) and methionine-induced VEI in rats. Results showed that HSAV significantly increased cell viability, inhibited apoptosis, and reduced the generation of reactive oxygen species (ROS) in H2O2-induced HUVECs. Meanwhile, HSAV decreased serum homocysteine (Hcy), endothelin 1 (ET-1), and oxidized low-density lipoprotein (ox-LDL) levels, increased nitric oxide (NO) and endothelin nitric oxide synthase (eNOS) levels, ameliorated pathological changes in rats with VEI induced by methionine. In parallel, HSAV relieved oxidative stress by decreasing malondialdehyde (MDA) level and increasing superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GSH-Px) levels in rats with VEI. Mechanism studies indicated that HSAV markedly downregulated the expression of protein kinase C zeta (PKCζ), and consequently regulated sirtuin 1 (Sirt1)-mediated oxidative stress signal pathway, and protein inhibitor of activated STATy (PIASy)-mediated apoptosis pathway, leading to the alleviation of oxidative stress and inhibition of apoptosis. These regulative effects of HSAV were further validated by knockdown and overexpression of PKCζ in vitro. In conclusion, HSAV showed protective effect against VEI by inhibiting PKCζ and, thereby, ameliorating oxidative stress and inhibiting apoptosis. This study not only provides guidance for the consumption of vinegar in daily life but also promotes the development of diet supplement for disease prevention.
Collapse
Affiliation(s)
- Xin Li
- Jiangsu Hengshun Vinegar Industry Co., Ltd., Zhenjiang, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Shenghu Zhu
- Jiangsu Hengshun Vinegar Industry Co., Ltd., Zhenjiang, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Bao Zhang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yongjian Yu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Jan M, Cueto R, Jiang X, Lu L, Sardy J, Xiong X, Yu JE, Pham H, Khan M, Qin X, Ji Y, Yang XF, Wang H. Molecular processes mediating hyperhomocysteinemia-induced metabolic reprogramming, redox regulation and growth inhibition in endothelial cells. Redox Biol 2021; 45:102018. [PMID: 34140262 PMCID: PMC8282538 DOI: 10.1016/j.redox.2021.102018] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/13/2021] [Accepted: 05/19/2021] [Indexed: 01/04/2023] Open
Abstract
Hyperhomocysteinemia (HHcy) is an established and potent independent risk factor for degenerative diseases, including cardiovascular disease (CVD), Alzheimer disease, type II diabetes mellitus, and chronic kidney disease. HHcy has been shown to inhibit proliferation and promote inflammatory responses in endothelial cells (EC), and impair endothelial function, a hallmark for vascular injury. However, metabolic processes and molecular mechanisms mediating HHcy-induced endothelial injury remains to be elucidated. This study examined the effects of HHcy on the expression of microRNA (miRNA) and mRNA in human aortic EC treated with a pathophysiologically relevant concentration of homocysteine (Hcy 500 μM). We performed a set of extensive bioinformatics analyses to identify HHcy-altered metabolic and molecular processes. The global functional implications and molecular network were determined by Gene Set Enrichment Analysis (GSEA) followed by Cytoscape analysis. We identified 244 significantly differentially expressed (SDE) mRNA, their relevant functional pathways, and 45 SDE miRNA. HHcy-altered SDE inversely correlated miRNA-mRNA pairs (45 induced/14 reduced mRNA) were discovered and applied to network construction using an experimentally verified database. We established a hypothetical model to describe the biochemical and molecular network with these specified miRNA/mRNA axes, finding: 1) HHcy causes metabolic reprogramming by increasing glucose uptake and oxidation, by glycogen debranching and NAD+/CoA synthesis, and by stimulating mitochondrial reactive oxygen species production via NNT/IDH2 suppression-induced NAD+/NADP-NADPH/NADP+ metabolism disruption; 2) HHcy activates inflammatory responses by activating inflammasome-pyroptosis mainly through ↓miR193b→↑CASP-9 signaling and by inducing IL-1β and adhesion molecules through the ↓miR29c→↑NEDD9 and the ↓miR1256→↑ICAM-1 axes, as well as GPCR and interferon α/β signaling; 3) HHcy promotes cell degradation by the activation of lysosome autophagy and ubiquitin proteasome systems; 4) HHcy causes cell cycle arrest at G1/S and S/G2 transitions, suppresses spindle checkpoint complex and cytokinetic abscission, and suppresses proliferation through ↓miRNA335/↑VASH1 and other axes. These findings are in accordance with our previous studies and add a wealth of heretofore-unexplored molecular and metabolic mechanisms underlying HHcy-induced endothelial injury. This is the first study to consider the effects of HHcy on both global mRNA and miRNA expression changes for mechanism identification. Molecular axes and biochemical processes identified in this study are useful not only for the understanding of mechanisms underlying HHcy-induced endothelial injury, but also for discovering therapeutic targets for CVD in general. Identified multiple HHcy-altered metabolic and molecular processes potentially responsible for HHcy-induced endothelial injury via examining global mRNA/miRNA expression changes in Hcy-treated EC and performing comprehensive bioinformatic studies. HHcy may activate glucose uptake signaling via the ↓miR148b→↑SLC2A axis. HHcy may induce glucose oxidation signaling by switching pyruvate metabolism from lactate synthesis to mitochondrial oxidation via expression changes of ↑MPC1 & ↓LDHB. HHcy may disrupt redox homeostasis mostly by suppressing NNT/IDH2-related mt-NADPH/mt-NAD+ signaling. HHcy may increase FA β-oxidation, glutamine, TCA cycle and OXPHOS signaling. HHcy may activate inflammatory signaling via the ↓miR29c→↑NEDD9 and the ↓miR1256→↑ICAM-1 axes. HHcy may activate inflammasome/pyroptosis-related signaling by the ↓miR137→↑TLR3, the ↓miR574→↑TRAF5, and the ↓miR193b→↑CASP-9 axes, and induce IL1α/β and CASP-10/7. HHcy may induce inflammation signaling via GPCR activation through the ↓miRNA335→↑CXCR4/↑GNA14 axes. HHcy may activate molecular degradation process signaling through the ↓miRNA335→↑ASAH1/↑ABCB9 axes. HHcy may suppress cell cycle and proliferation through the miR491→↓HMGA2→↓CCNA2/CCNB2, the ↓miR335→↑VASH1, the ↓miR181a→↑PHLDA1, the miR6045→↓CENPH, the miR22→↓PRR11/↓BRCA2, and the miR605/miR497/miR514a→CEP55 axes
Collapse
Affiliation(s)
- Michael Jan
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, United States
| | - Ramon Cueto
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Liu Lu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Jason Sardy
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xinyu Xiong
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Justine E Yu
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Hung Pham
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States
| | - Xuebing Qin
- Tulane National Primate Research Center, School of Medicine, Tulane University, Covington, LA, United States
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Xiao-Feng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, United States
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, Philadelphia, PA, United States; Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
18
|
Moretti R, Giuffré M, Caruso P, Gazzin S, Tiribelli C. Homocysteine in Neurology: A Possible Contributing Factor to Small Vessel Disease. Int J Mol Sci 2021; 22:2051. [PMID: 33669577 PMCID: PMC7922986 DOI: 10.3390/ijms22042051] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Homocysteine (Hcy) is a sulfur-containing amino acid generated during methionine metabolism, accumulation of which may be caused by genetic defects or the deficit of vitamin B12 and folate. A serum level greater than 15 micro-mols/L is defined as hyperhomocysteinemia (HHcy). Hcy has many roles, the most important being the active participation in the transmethylation reactions, fundamental for the brain. Many studies focused on the role of homocysteine accumulation in vascular or degenerative neurological diseases, but the results are still undefined. More is known in cardiovascular disease. HHcy is a determinant for the development and progression of inflammation, atherosclerotic plaque formation, endothelium, arteriolar damage, smooth muscle cell proliferation, and altered-oxidative stress response. Conversely, few studies focused on the relationship between HHcy and small vessel disease (SVD), despite the evidence that mice with HHcy showed a significant end-feet disruption of astrocytes with a diffuse SVD. A severe reduction of vascular aquaporin-4-water channels, lower levels of high-functioning potassium channels, and higher metalloproteinases are also observed. HHcy modulates the N-homocysteinylation process, promoting a pro-coagulative state and damage of the cellular protein integrity. This altered process could be directly involved in the altered endothelium activation, typical of SVD and protein quality, inhibiting the ubiquitin-proteasome system control. HHcy also promotes a constant enhancement of microglia activation, inducing the sustained pro-inflammatory status observed in SVD. This review article addresses the possible role of HHcy in small-vessel disease and understands its pathogenic impact.
Collapse
Affiliation(s)
- Rita Moretti
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Mauro Giuffré
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Paola Caruso
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Silvia Gazzin
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| | - Claudio Tiribelli
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| |
Collapse
|
19
|
Neutrophil counts and the risk of first stroke in general hypertensive adults. Hypertens Res 2021; 44:830-839. [PMID: 33564178 DOI: 10.1038/s41440-021-00625-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 11/09/2022]
Abstract
We aimed to investigate the association between neutrophil counts and first stroke and examine possible effect modifiers among treated hypertensive adults. This is a post hoc analysis of the China Stroke Primary Prevention Trial (CSPPT). A total of 11,878 hypertensive adults with data on neutrophil counts at baseline were included in the current study. The primary outcome was first stroke. During a median follow-up of 4.5 years, 414 (3.5%) participants experienced a first stroke, including 358 with ischemic stroke, 55 with hemorrhagic stroke and one with uncertain type of stroke. Compared with participants in quartile 1 (<2.9 × 109/L) of neutrophil counts, those in the upper quartiles (quartile 2-4 [≥2.9 × 109/L]) had a significantly higher risk of first stroke (HR, 1.35; 95% CI: 1.02, 1.78) or first ischemic stroke (HR, 1.38; 95% CI: 1.02, 1.86). Moreover, a strong positive association between neutrophil counts and first ischemic stroke was found in participants with total homocysteine (tHcy) levels <15 μmol/L (HR, 1.74; 95% CI: 1.17, 2.58; vs. ≥15 μmol/L; HR, 0.91; 95% CI: 0.57, 1.46, P interaction = 0.042) at baseline or time-averaged mean arterial pressure (MAP) ≥102 mmHg (median) (HR, 1.92; 95% CI: 1.27, 2.89; vs. <102 mmHg; HR, 0.89; 95% CI: 0.57, 1.41, P interaction = 0.015) during the treatment period. However, no such association between neutrophil counts and first hemorrhagic stroke was found. In summary, high baseline neutrophil counts were associated with an increased risk of first ischemic stroke among hypertensive patients, especially in those with low tHcy at baseline or high time-averaged MAP during the treatment period.
Collapse
|
20
|
Lian N, Shi LQ, Hao ZM, Chen M. Research progress and perspective in metabolism and metabolomics of psoriasis. Chin Med J (Engl) 2020; 133:2976-2986. [PMID: 33237698 PMCID: PMC7752687 DOI: 10.1097/cm9.0000000000001242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT Psoriasis is considered a systemic disease associated with metabolic abnormalities, and it is important to understand the mechanisms by which metabolism affects pathophysiological processes both holistically and systematically. Metabolites are closely related to disease phenotypes, especially in systemic diseases under multifactorial modulation. The emergence of metabolomics has provided information regarding metabolite changes in lesions and circulation and deepened our understanding of the association between metabolic reprogramming and psoriasis. Metabolomics has great potential for the development of effective biomarkers for clinical diagnosis, therapeutic monitoring, prediction of the efficacy of psoriasis management, and further discovery of new metabolism-based therapeutic targets.
Collapse
Affiliation(s)
- Ni Lian
- Department of Dermatology, Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences & Peking Union Medical Collage, Nanjing, Jiangsu 210042, China
| | | | | | | |
Collapse
|
21
|
HMGB1 mediates homocysteine-induced endothelial cells pyroptosis via cathepsin V-dependent pathway. Biochem Biophys Res Commun 2020; 532:640-646. [DOI: 10.1016/j.bbrc.2020.08.091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 08/24/2020] [Indexed: 01/20/2023]
|
22
|
Ma C, Zhao Y, Liu Z. Vitamin D Provides Benefit Based on the Proinflammatory Effects of Homocysteine in Elderly Patients With Type 2 Diabetes Mellitus. Clin Ther 2020; 42:2010-2020.e1. [PMID: 32921500 DOI: 10.1016/j.clinthera.2020.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/28/2020] [Accepted: 08/16/2020] [Indexed: 12/09/2022]
Abstract
PURPOSE It is unclear whether vitamin D provides any benefit against the pro-inflammatory effects of homocysteine in elderly patients with type 2 diabetes mellitus (T2DM). METHODS We compared lymphocyte counts for CD3, CD19, CD4, and CD8 subsets between elderly (age ≥65 years) T2DM patients (n = 5098) and nondiabetes control subjects (n = 20,590) based on the serum concentrations of homocysteine and total vitamin D (calcidiol + calcifediol [total vitamin D, TVD]; <20, 20-30, and >30 ng/mL). FINDINGS Significant variation in CD19 (P = 0.019), CD4 (P = 0.015), and CD8 (P < 0.001) were associated with serum TVD in T2DM patients with homocysteine ≤15 μmol/L, whereas CD3 (P = 0.003) and CD8 (P = 0.019) varied in control subjects with homocysteine ≤15 μmol/L. In T2DM patients with high homocysteine (>15 μmol/L) levels, significant variation based on serum TVD occurred in CD19 only (P = 0.024), whereas CD3 (P = 0.016) and CD4 (P = 0.001) varied in control subjects with high homocysteine concentrations. IMPLICATIONS Serum TVD influences variation in CD3, CD19, CD4, and CD8 lymphocyte subsets based on the serum homocysteine concentration in elderly T2DM patients and nondiabetic individuals with moderate to high homocysteine concentrations. The effect of TVD is partially attenuated in individuals with high homocysteine concentrations, with greater attenuation occurring in patients with T2DM. Differences in the variation of lymphocyte subsets between nondiabetes subjects with moderate homocysteine concentrations and those with high homocysteine concentrations constitute a shift from CD8-positive cells to CD4-positive cells, suggesting a change in TH1/TH2 balance based on TVD and homocysteine concentrations that is absent in diabetes cases with high homocysteine concentrations.
Collapse
Affiliation(s)
- Cong Ma
- Department of Endocrinology, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Yinxia Zhao
- Central Laboratory, Shanghai Xuhui Central Hospital, Shanghai, China
| | - Zhiwen Liu
- Department of Endocrinology, Shanghai Xuhui Central Hospital, Shanghai, China.
| |
Collapse
|
23
|
Clotet-Freixas S, McEvoy CM, Batruch I, Pastrello C, Kotlyar M, Van JAD, Arambewela M, Boshart A, Farkona S, Niu Y, Li Y, Famure O, Bozovic A, Kulasingam V, Chen P, Kim SJ, Chan E, Moshkelgosha S, Rahman SA, Das J, Martinu T, Juvet S, Jurisica I, Chruscinski A, John R, Konvalinka A. Extracellular Matrix Injury of Kidney Allografts in Antibody-Mediated Rejection: A Proteomics Study. J Am Soc Nephrol 2020; 31:2705-2724. [PMID: 32900843 DOI: 10.1681/asn.2020030286] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Antibody-mediated rejection (AMR) accounts for >50% of kidney allograft loss. Donor-specific antibodies (DSA) against HLA and non-HLA antigens in the glomeruli and the tubulointerstitium cause AMR while inflammatory cytokines such as TNFα trigger graft injury. The mechanisms governing cell-specific injury in AMR remain unclear. METHODS Unbiased proteomic analysis of laser-captured and microdissected glomeruli and tubulointerstitium was performed on 30 for-cause kidney biopsy specimens with early AMR, acute cellular rejection (ACR), or acute tubular necrosis (ATN). RESULTS A total of 107 of 2026 glomerular and 112 of 2399 tubulointerstitial proteins was significantly differentially expressed in AMR versus ACR; 112 of 2026 glomerular and 181 of 2399 tubulointerstitial proteins were significantly dysregulated in AMR versus ATN (P<0.05). Basement membrane and extracellular matrix (ECM) proteins were significantly decreased in both AMR compartments. Glomerular and tubulointerstitial laminin subunit γ-1 (LAMC1) expression decreased in AMR, as did glomerular nephrin (NPHS1) and receptor-type tyrosine-phosphatase O (PTPRO). The proteomic analysis revealed upregulated galectin-1, which is an immunomodulatory protein linked to the ECM, in AMR glomeruli. Anti-HLA class I antibodies significantly increased cathepsin-V (CTSV) expression and galectin-1 expression and secretion in human glomerular endothelial cells. CTSV had been predicted to cleave ECM proteins in the AMR glomeruli. Glutathione S-transferase ω-1, an ECM-modifying enzyme, was significantly increased in the AMR tubulointerstitium and in TNFα-treated proximal tubular epithelial cells. CONCLUSIONS Basement membranes are often remodeled in chronic AMR. Proteomic analysis performed on laser-captured and microdissected glomeruli and tubulointerstitium identified early ECM remodeling, which may represent a new therapeutic opportunity.
Collapse
Affiliation(s)
- Sergi Clotet-Freixas
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Caitriona M McEvoy
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Ihor Batruch
- Department of Laboratory Medicine and Pathobiology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Chiara Pastrello
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Max Kotlyar
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Julie Anh Dung Van
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Madhurangi Arambewela
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Alex Boshart
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Farkona
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yun Niu
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Yanhong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Olusegun Famure
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Andrea Bozovic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Vathany Kulasingam
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Peixuen Chen
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - S Joseph Kim
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Emilie Chan
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Sajad Moshkelgosha
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Syed Ashiqur Rahman
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Center for Systems Immunology, Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jishnu Das
- Center for Systems Immunology, Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Center for Systems Immunology, Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tereza Martinu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Respirology, Toronto Lung Transplant Program, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Andrzej Chruscinski
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| | - Rohan John
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ana Konvalinka
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada .,Division of Nephrology, Department of Medicine, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Soham and Shaila Ajmera Family Transplant Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Serum Protein Changes in Pediatric Sepsis Patients Identified With an Aptamer-Based Multiplexed Proteomic Approach. Crit Care Med 2020; 48:e48-e57. [PMID: 31714400 DOI: 10.1097/ccm.0000000000004083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Sepsis, a life-threatening organ dysfunction caused by a dysregulated host response to infection, is a leading cause of death and disability among children worldwide. Identifying sepsis in pediatric patients is difficult and can lead to treatment delay. Our objective was to assess the host proteomic response to infection utilizing an aptamer-based multiplexed proteomics approach to identify novel serum protein changes that might help distinguish between pediatric sepsis and infection-negative systemic inflammation and hence can potentially improve sensitivity and specificity of the diagnosis of sepsis over current clinical criteria approaches. DESIGN Retrospective, observational cohort study. SETTING PICU and cardiac ICU, Seattle Children's Hospital, Seattle, WA. PATIENTS A cohort of 40 children with clinically overt sepsis and 30 children immediately postcardiopulmonary bypass surgery (infection-negative systemic inflammation control subjects) was recruited. Children with sepsis had a confirmed or suspected infection, two or more systemic inflammatory response syndrome criteria, and at least cardiovascular and/or pulmonary organ dysfunction. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Serum samples from 35 of the sepsis and 28 of the bypass surgery subjects were available for screening with an aptamer-based proteomic platform that measures 1,305 proteins to search for large-scale serum protein expression pattern changes in sepsis. A total of 111 proteins were significantly differentially expressed between the sepsis and control groups, using the linear models for microarray data (linear modeling) and Boruta (decision trees) R packages, with 55 being previously identified in sepsis patients. Weighted gene correlation network analysis helped identify 76 proteins that correlated highly with clinical sepsis traits, 27 of which had not been previously reported in sepsis. CONCLUSIONS The serum protein changes identified with the aptamer-based multiplexed proteomics approach used in this study can be useful to distinguish between sepsis and noninfectious systemic inflammation.
Collapse
|
25
|
De Pasquale V, Moles A, Pavone LM. Cathepsins in the Pathophysiology of Mucopolysaccharidoses: New Perspectives for Therapy. Cells 2020; 9:cells9040979. [PMID: 32326609 PMCID: PMC7227001 DOI: 10.3390/cells9040979] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cathepsins (CTSs) are ubiquitously expressed proteases normally found in the endolysosomal compartment where they mediate protein degradation and turnover. However, CTSs are also found in the cytoplasm, nucleus, and extracellular matrix where they actively participate in cell signaling, protein processing, and trafficking through the plasma and nuclear membranes and between intracellular organelles. Dysregulation in CTS expression and/or activity disrupts cellular homeostasis, thus contributing to many human diseases, including inflammatory and cardiovascular diseases, neurodegenerative disorders, diabetes, obesity, cancer, kidney dysfunction, and others. This review aimed to highlight the involvement of CTSs in inherited lysosomal storage disorders, with a primary focus to the emerging evidence on the role of CTSs in the pathophysiology of Mucopolysaccharidoses (MPSs). These latter diseases are characterized by severe neurological, skeletal and cardiovascular phenotypes, and no effective cure exists to date. The advance in the knowledge of the molecular mechanisms underlying the activity of CTSs in MPSs may open a new challenge for the development of novel therapeutic approaches for the cure of such intractable diseases.
Collapse
Affiliation(s)
- Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish Research Council, 08036 Barcelona, Spain;
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
- Correspondence: ; Tel.: +39-081-7463043
| |
Collapse
|
26
|
Wei M, Wang L, Liu YS, Zheng MQ, Ma FF, Qi YC, Liu G. Homocysteine as a potential predictive factor for high major adverse cardiovascular events risk in female patients with premature acute coronary syndrome. Medicine (Baltimore) 2019; 98:e18019. [PMID: 31764817 PMCID: PMC6882653 DOI: 10.1097/md.0000000000018019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We aimed to investigate the correlation of homocysteine (Hcy) level with clinical characteristics, and explore its predictive value for major adverse cardiovascular events (MACE) risk in female patients with premature acute coronary syndrome (ACS).The serum Hcy level was detected from 1299 female patients with premature ACS. According to the tertile of Hcy level, patients were divided into 3 groups: lowest tertile group (≤9.1 μmol/L), middle tertile group (9.2-11.6 μmol/L) and highest tertile group (>11.6 μmol/L). MACE incidence was recorded and MACE-free survival was caculated with the median follow-up duration of 28.3 months.Increased Hcy correlated with older age (P < .001), higher creatinine level (P < .001), and enhanced uric acid level (P = .001), while reduced fasting glucose concentration (P < .001). MACE incidence was 10.7% and it was highest in highest tertile group (22.1%), followed by middle tertile group (7.7%) and lowest tertile group (2.4%) (P < .001). Receiver operating characteristic curve showed that Hcy distinguished MACE patients from non-MACE patients with the area under the curve of 0.789 (95% CI: 0.742-0.835). Kaplan-Meier curves revealed that MACE-free survival was shortest in Hcy highest tertile group, followed by middle tertile group and lowest tertile group (P < .001). Multivariate Cox analyses further showed that higher Hcy level was an independent predictive factor for poor MACE-free survival (middle tertile vs lowest tertile (P = .001, HR: 3.615, 95% CI: 1.661-7.864); highest tertile vs lowest tertile (P < .001, HR: 11.023, 95% CI: 5.356-22.684)).Hcy serves as a potential predictive factor for increased MACE risk in female patients with premature ACS.
Collapse
Affiliation(s)
- Mei Wei
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang
| | - Le Wang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin
| | - Yong-Sheng Liu
- Department of General Family Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ming-Qi Zheng
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang
| | - Fang-Fang Ma
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang
| | - Yan-Chao Qi
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang
| | - Gang Liu
- Heart Center, The First Hospital of Hebei Medical University, Shijiazhuang
| |
Collapse
|
27
|
Si J, Li XW, Wang Y, Zhang YH, Wu QQ, Zhang LM, Zuo XB, Gao J, Li J. Relationship between serum homocysteine levels and long-term outcomes in patients with ST-segment elevation myocardial infarction. Chin Med J (Engl) 2019; 132:1028-1036. [PMID: 30829711 PMCID: PMC6595884 DOI: 10.1097/cm9.0000000000000159] [Citation(s) in RCA: 2] [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: 11/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The mortality of cardiovascular disease is constantly rising, and novel biomarkers help us predict residual risk. This study aimed to evaluate the predictive value of serum homocysteine (HCY) levels on prognosis in patients with ST-segment elevation myocardial infarction (STEMI). METHODS The 419 consecutive patients with STEMI, treated at one medical center, from March 2010 to December 2015 were retrospectively investigated. Peripheral blood samples were obtained within 24 h of admission and HCY concentrations were measured using an enzymatic cycling assay. The patients were divided into high HCY level (H-HCY) and low HCY level (L-HCY) groups. Short- and long-term outcomes were compared, as were age-based subgroups (patients aged 60 years and younger vs. those older than 60 years). Statistical analyses were mainly conducted by Student t-test, Chi-squared test, logistic regression, and Cox proportional-hazards regression. RESULTS The H-HCY group had more males (84.6% vs. 75.4%, P = 0.018), and a lower prevalence of diabetes (20.2% vs. 35.5%, P < 0.001), compared with the L-HCY group. During hospitalization, there were seven mortalities in the L-HCY group and 10 in the H-HCY group (3.3% vs. 4.8%, P = 0.440). During the median follow-up period of 35.8 (26.9-46.1) months, 33 (16.2%) patients in the L-HCY group and 48 (24.2%) in the H-HCY group experienced major adverse cardiovascular and cerebrovascular events (MACCE) (P = 0.120). History of hypertension (hazard ratio [HR]: 1.881, 95% confidence interval [CI]: 1.178-3.005, P = 0.008) and higher Killip class (HR: 1.923, 95% CI: 1.419-2.607, P < 0.001), but not HCY levels (HR: 1.007, 95% CI: 0.987-1.027, P = 0.507), were significantly associated with long-term outcomes. However, the subgroup analysis indicated that in older patients, HCY levels were significantly associated with long-term outcomes (HR: 1.036, 95% CI: 1.011-1.062, P = 0.005). CONCLUSION Serum HCY levels did not independently predict in-hospital or long-term outcomes in patients with STEMI; however, among elderly patients with STEMI, this study revealed a risk profile for late outcomes that incorporated HCY level.
Collapse
Affiliation(s)
- Jin Si
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xue-Wen Li
- Department of Cardiology, Tianjin Medical University, Tianjin 300070, China
- Cadre's Ward, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin 300162, China
| | - Yang Wang
- Medical Research & Biometrics Center, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Ying-Hua Zhang
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qing-Qing Wu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Lei-Min Zhang
- Department of Cardiology, Qinghe Substation Hospital of Beijing Municipal Administration of Prisons, Tianjin 300481, China
| | - Xue-Bing Zuo
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing Gao
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing Li
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
28
|
Ma Y, Zhang Z, Chen R, Shi R, Zeng P, Chen R, Leng Y, Chen AF. NRP1 regulates HMGB1 in vascular endothelial cells under high homocysteine condition. Am J Physiol Heart Circ Physiol 2019; 316:H1039-H1046. [PMID: 30767669 DOI: 10.1152/ajpheart.00746.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial inflammation plays an important role in hyperhomocysteinemia (HHcy)-associated vascular diseases. High mobility group box 1 (HMGB1) is a pro-inflammatory danger molecule produced by endothelial cells. However, whether HMGB1 is involved in vascular endothelial inflammation of HHcy is poorly understood. Neuropilin-1 (NRP1) mediates inflammatory response and activates mitogen-activated protein kinases (MAPKs) pathway that has been reported to be involved in regulation of HMGB1. The aim of this study was to determine the alteration of HMGB1 in HHcy, and the role of NRP1 in regulation of endothelial HMGB1 under high homocysteine (Hcy) condition. In the present study, we first observed that the plasma level of HMGB1 was elevated in HHcy patients and an experimental rat model, and increased HMGB1 was also observed in the thoracic aorta of an HHcy rat model. HMGB1 was induced by Hcy accompanied with upregulated NRP1 in vascular endothelial cells. Overexpression of NRP1 promoted expression and secretion of HMGB1 and endothelial inflammation; knockdown of NRP1 inhibited HMGB1 and endothelial inflammation induced by Hcy, which partially regulated through p38 MAPK pathway. Furthermore, NRP1 inhibitor ATWLPPR reduced plasma HMGB1 level and expression of HMGB1 in the thoracic aorta of HHcy rats. In conclusion, our data suggested that Hcy requires NRP1 to regulate expression and secretion of HMGB1. The present study provides the evidence for inhibition of NRP1 and HMGB1 to be the novel therapeutic targets of vascular endothelial inflammation in HHcy in the future. NEW & NOTEWORTHY This study shows for the first time to our knowledge that the plasma level of high mobility group box 1 (HMGB1) is elevated in hyperhomocysteinemia (HHcy) patients, and homocysteine promotes expression and secretion of HMGB1 partially regulated by neuropilin-1 in endothelial cells, which is involved in endothelial inflammation. Most importantly, these new findings will provide a potential therapeutic strategy for vascular endothelial inflammation in HHcy.
Collapse
Affiliation(s)
- Yeshuo Ma
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Zhen Zhang
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Runtai Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Rui Shi
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Xiangya School of Pharmaceutical Sciences, Central South University , Changsha , China
| | - Pingyu Zeng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Ruifang Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Yiping Leng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| | - Alex F Chen
- Department of Cardiology, The Third Xiangya Hospital of Central South University , Changsha , China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University , Changsha , China
| |
Collapse
|
29
|
He R, Yan X, Guo J, Xu Q, Tang B, Sun Q. Recent Advances in Biomarkers for Parkinson's Disease. Front Aging Neurosci 2018; 10:305. [PMID: 30364199 PMCID: PMC6193101 DOI: 10.3389/fnagi.2018.00305] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 09/14/2018] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is one of the common progressive neurodegenerative disorders with several motor and non-motor symptoms. Most of the motor symptoms may appear at a late stage where most of the dopaminergic neurons have been already damaged. In order to provide better clinical intervention and treatment at the onset of disease, it is imperative to find accurate biomarkers for early diagnosis, including prodromal diagnosis and preclinical diagnosis. At the same time, these reliable biomarkers can also be utilized to monitor the progress of the disease. In this review article, we will discuss recent advances in the development of PD biomarkers from different aspects, including clinical, biochemical, neuroimaging and genetic aspects. Although various biomarkers for PD have been developed so far, their specificity and sensitivity are not ideal when applied individually. So, the combination of multimodal biomarkers will greatly improve the diagnostic accuracy and facilitate the implementation of personalized medicine.
Collapse
Affiliation(s)
- Runcheng He
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxiang Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
| | - Qian Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- Parkinson’s Disease Center of Beijing Institute for Brain Disorders, Beijing, China
- Collaborative Innovation Center for Brain Science, Shanghai, China
- Collaborative Innovation Center for Genetics and Development, Shanghai, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qiying Sun
- National Clinical Research Center for Geriatric Disorders, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Luo Z, Lu Z, Muhammad I, Chen Y, Chen Q, Zhang J, Song Y. Associations of the MTHFR rs1801133 polymorphism with coronary artery disease and lipid levels: a systematic review and updated meta-analysis. Lipids Health Dis 2018; 17:191. [PMID: 30115070 PMCID: PMC6097444 DOI: 10.1186/s12944-018-0837-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The associations of the 5,10-methylenetetrahydrofolate reductase gene (MTHFR) rs1801133 polymorphism with coronary artery disease (CAD) and plasma lipid levels have been widely investigated, but the results were inconsistent and inconclusive. This meta-analysis aimed to clarify the relationships of the rs1801133 polymorphism with CAD and plasma lipid levels. METHODS By searching in PubMed, Google Scholar, Web of Science, Cochrane Library, Wanfang, VIP and CNKI databases, 123 studies (87,020 subjects) and 65 studies (85,554 subjects) were identified for the CAD association analysis and the lipid association analysis, respectively. Odds ratio (OR) and standardized mean difference (SMD) were used to determine the effects of the rs1801133 polymorphism on CAD risk and lipid levels, respectively. RESULTS The variant T allele of the rs1801133 polymorphism was associated with increased risk of CAD under allelic model [OR = 1.11, 95% confidence interval (CI) = 1.06-1.17, P < 0.01], additive model (OR = 1.25, 95% CI = 1.14-1.37, P < 0.01), dominant model (OR = 1.11, 95% CI = 1.04-1.17, P < 0.01), and recessive model (OR = 1.22, 95% CI = 1.12-1.32, P < 0.01). The T carriers had higher levels of total cholesterol (TC) (SMD = 0.04, 95% CI = 0.01-0.07, P = 0.02) and low-density lipoprotein cholesterol (LDL-C) (SMD = 0.07, 95% CI = 0.01-0.12, P = 0.01) than the non-carriers. CONCLUSIONS The meta-analysis suggested that the T allele of the rs1801133 polymorphism is a risk factor for CAD, which is possibly and partly mediated by abnormal lipid levels.
Collapse
Affiliation(s)
- Zhi Luo
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Zhan Lu
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Irfan Muhammad
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yun Chen
- Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Qiuhong Chen
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Jiaojiao Zhang
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Yongyan Song
- Department of Medical Biochemistry, School of Preclinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
| |
Collapse
|
31
|
Wang X, Huang Y, Ji Y. Spotlight on small molecules in cardiovascular diseases. Br J Pharmacol 2018; 175:1111-1113. [PMID: 29574891 PMCID: PMC5866956 DOI: 10.1111/bph.14154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Xin Wang
- Faculty of Life SciencesThe University of ManchesterManchesterUK
| | - Yu Huang
- Institute of Vascular BiologyChinese University of Hong KongSha TinHong Kong
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of PharmacyNanjing Medical UniversityNanjingChina
| |
Collapse
|
32
|
Cocchiaro P, De Pasquale V, Della Morte R, Tafuri S, Avallone L, Pizard A, Moles A, Pavone LM. The Multifaceted Role of the Lysosomal Protease Cathepsins in Kidney Disease. Front Cell Dev Biol 2017; 5:114. [PMID: 29312937 PMCID: PMC5742100 DOI: 10.3389/fcell.2017.00114] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/07/2017] [Indexed: 12/18/2022] Open
Abstract
Kidney disease is worldwide the 12th leading cause of death affecting 8–16% of the entire population. Kidney disease encompasses acute (short-lasting episode) and chronic (developing over years) pathologies both leading to renal failure. Since specific treatments for acute or chronic kidney disease are limited, more than 2 million people a year require dialysis or kidney transplantation. Several recent evidences identified lysosomal proteases cathepsins as key players in kidney pathophysiology. Cathepsins, originally found in the lysosomes, exert important functions also in the cytosol and nucleus of cells as well as in the extracellular space, thus participating in a wide range of physiological and pathological processes. Based on their catalytic active site residue, the 15 human cathepsins identified up to now are classified in three different families: serine (cathepsins A and G), aspartate (cathepsins D and E), or cysteine (cathepsins B, C, F, H, K, L, O, S, V, X, and W) proteases. Specifically in the kidney, cathepsins B, D, L and S have been shown to regulate extracellular matrix homeostasis, autophagy, apoptosis, glomerular permeability, endothelial function, and inflammation. Dysregulation of their expression/activity has been associated to the onset and progression of kidney disease. This review summarizes most of the recent findings that highlight the critical role of cathepsins in kidney disease development and progression. A better understanding of the signaling pathways governed by cathepsins in kidney physiopathology may yield novel selective biomarkers or therapeutic targets for developing specific treatments against kidney disease.
Collapse
Affiliation(s)
- Pasquale Cocchiaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,Faculty of Medicine, Institut National de la Santé Et de la Recherche Médicale, "Défaillance Cardiaque Aigüe et Chronique", Nancy, France.,Université de Lorraine, Nancy, France.,Institut Lorrain du Coeur et des Vaisseaux, Center for Clinical Investigation 1433, Nancy, France.,CHRU de Nancy, Hôpitaux de Brabois, Nancy, France
| | - Valeria De Pasquale
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Rossella Della Morte
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Simona Tafuri
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luigi Avallone
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Anne Pizard
- Faculty of Medicine, Institut National de la Santé Et de la Recherche Médicale, "Défaillance Cardiaque Aigüe et Chronique", Nancy, France.,Université de Lorraine, Nancy, France.,Institut Lorrain du Coeur et des Vaisseaux, Center for Clinical Investigation 1433, Nancy, France.,CHRU de Nancy, Hôpitaux de Brabois, Nancy, France
| | - Anna Moles
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
33
|
Leng YP, Ma YS, Li XG, Chen RF, Zeng PY, Li XH, Qiu CF, Li YP, Zhang Z, Chen AF. l-Homocysteine-induced cathepsin V mediates the vascular endothelial inflammation in hyperhomocysteinaemia. Br J Pharmacol 2017. [PMID: 28631302 DOI: 10.1111/bph.13920] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Vascular inflammation, including the expression of inflammatory cytokines in endothelial cells, plays a critical role in hyperhomocysteinaemia-associated vascular diseases. Cathepsin V, specifically expressed in humans, is involved in vascular diseases through its elastolytic and collagenolytic activities. The aim of this study was to determine the effects of cathepsin V on l-homocysteine-induced vascular inflammation. EXPERIMENTAL APPROACH A high methionine diet-induced hyperhomocysteinaemic mouse model was used to assess cathepsin V expression and vascular inflammation. Cultures of HUVECs were challenged with l-homocysteine and the cathepsin L/V inhibitor SID to assess the pro-inflammatory effects of cathepsin V. Transfection and antisense techniques were utilized to investigate the effects of cathepsin V on the dual-specificity protein phosphatases (DUSPs) and MAPK pathways. KEY RESULTS Cathepsin L (human cathepsin V homologous) was increased in the thoracic aorta endothelial cells of hyperhomocysteinaemic mice; l-homocysteine promoted cathepsin V expression in HUVECs. SID suppressed the activity of cathepsin V and reversed the up-regulation of inflammatory cytokines (IL-6, IL-8 and TNF-α), adhesion and chemotaxis of leukocytes and vascular inflammation induced by l-homocysteine in vivo and in vitro. Increased cathepsin V promoted the degradation of DUSP6 and DUSP7, phosphorylation and subsequent nuclear translocation of ERK1/2, phosphorylation of STAT1 and expression of IL-6, IL-8 and TNF-α. CONCLUSIONS AND IMPLICATIONS This study has identified a novel mechanism, which shows that l-homocysteine-induced upregulation of cathepsin V mediates vascular endothelial inflammation under high homocysteine condition partly via ERK1/2 /STAT1 pathway. This mechanism could represent a potential therapeutic target in hyperaemia-associated vascular diseases. LINKED ARTICLES This article is part of a themed section on Spotlight on Small Molecules in Cardiovascular Diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.8/issuetoc.
Collapse
Affiliation(s)
- Yi-Ping Leng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ye-Shuo Ma
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiao-Gang Li
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Rui-Fang Chen
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ping-Yu Zeng
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiao-Hui Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Cheng-Feng Qiu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Ya-Pei Li
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Zhang
- Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China.,Centre for Experimental Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Alex F Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.,Center for Vascular Disease and Translational Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|