151
|
Filipová M, Bojarová P, Rodrigues Tavares M, Bumba L, Elling L, Chytil P, Gunár K, Křen V, Etrych T, Janoušková O. Glycopolymers for Efficient Inhibition of Galectin-3: In Vitro Proof of Efficacy Using Suppression of T Lymphocyte Apoptosis and Tumor Cell Migration. Biomacromolecules 2020; 21:3122-3133. [PMID: 32697592 DOI: 10.1021/acs.biomac.0c00515] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The development of efficient galectin-3 (Gal-3) inhibitors draws attention in the field of anti-cancer therapy, especially due to the prominent role of extra- and intracellular Gal-3 in vital processes of cancerogenesis, such as immunosuppression, stimulation of tumor cells proliferation, survival, invasion, apoptotic resistance, and metastasis formation and progression. Here, by combining poly-LacNAc (Galβ4GlcNAc)-derived oligosaccharides with N-(2-hydroxypropyl) methacrylamide (HPMA) copolymers, we synthesized multivalent glycopolymer inhibitors with a high potential to target extracellular and intracellular Gal-3. The inhibitory capabilities of the best conjugate in the studied series were in the nanomolar range proving the excellent Gal-3 inhibitory potential. Moreover, thorough investigation of the inhibitory effect in the biological conditions showed that the glycopolymers strongly inhibited Gal-3-induced apoptosis of T lymphocytes and suppressed migration and spreading of colorectal, breast, melanoma, and prostate cancer cells. In sum, the strong inhibitory activity toward Gal-3, combined with favorable pharmacokinetics of HPMA copolymers ensuring enhanced tumor accumulation via the enhanced permeability and retention effect, nominate the glycopolymers containing LacdiNAc-LacNAc (GalNAcβ4GlcNAcβ3Galβ4GlcNAc) tetrasaccharide as promising tools for preclinical in anti-cancer therapy evaluation.
Collapse
Affiliation(s)
- Marcela Filipová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic.,Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Sítná Sq. 3105, CZ-27201 Kladno, Czech Republic
| | - Marina Rodrigues Tavares
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic
| | - Lothar Elling
- Institute of Biotechnology and Helmholtz Institute for Biomedical Engineering, RWTH Aachen, Pauwelstr. 20, D-52079 Aachen, Germany
| | - Petr Chytil
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Kristýna Gunár
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4 Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského nám. 2, CZ-16206 Prague 6, Czech Republic
| |
Collapse
|
152
|
Vašíček T, Spiwok V, Červený J, Petrásková L, Bumba L, Vrbata D, Pelantová H, Křen V, Bojarová P. Regioselective 3-O-Substitution of Unprotected Thiodigalactosides: Direct Route to Galectin Inhibitors. Chemistry 2020; 26:9620-9631. [PMID: 32368810 DOI: 10.1002/chem.202002084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/26/2022]
Abstract
The synthesis of tailored bioactive carbohydrates usually comprises challenging (de)protection steps, which lowers synthetic yields and increases time demands. We present here a regioselective single-step introduction of benzylic substituents at 3-hydroxy groups of β-d-galactopyranosyl-(1→1)-thio-β-d-galactopyranoside (TDG) employing dibutyltin oxide in good yields. These glycomimetics act as inhibitors of galectins-human lectins, which are biomedically attractive targets for therapeutic inhibition in, for example, cancerogenesis. The affinity of the prepared glycomimetics to galectin-1 and galectin-3 was studied in enzyme-linked immunosorbent (ELISA)-type assays and their potential to inhibit galectin binding on the cell surface was shown. We used our original in vivo biotinylated galectin constructs for easy detection by flow cytometry. The results of the biological experiments were compared with data from molecular modeling with both galectins. The present work reveals a facile and elegant synthetic route for the preparation of TDG-derived glycomimetics that exhibit differing selectivity and affinity to galectins depending on the choice of 3-O-substitution.
Collapse
Affiliation(s)
- Tomáš Vašíček
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Vojtěch Spiwok
- University of Chemistry and Technology Prague, Technická 3, 16628, Prague 6, Czech Republic
| | - Jakub Červený
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Lucie Petrásková
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Ladislav Bumba
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - David Vrbata
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Helena Pelantová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Vladimír Křen
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic
| | - Pavla Bojarová
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Vídeňská 1083, 14220, Prague 4, Czech Republic.,Department of Health Care Disciplines and Population Protection, Faculty of Biomedical Engineering, Czech Technical University in Prague, Nám. Sítná 3105, 27201, Kladno, Czech Republic
| |
Collapse
|
153
|
Aguilar D, Sun C, Hoogeveen RC, Nambi V, Selvin E, Matsushita K, Saeed A, McEvoy JW, Shah AM, Solomon SD, Boerwinkle E, Ballantyne CM. Levels and Change in Galectin-3 and Association With Cardiovascular Events: The ARIC Study. J Am Heart Assoc 2020; 9:e015405. [PMID: 32573308 PMCID: PMC7670497 DOI: 10.1161/jaha.119.015405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Background Circulating galectin‐3 levels provide prognostic information in patients with established heart failure (HF), but the associations between galectin‐3 levels and other incident cardiovascular events in asymptomatic individuals at midlife and when remeasured ≈15 years later are largely uncharacterized. Methods and Results Using multivariable Cox proportional hazards models, we identified associations between plasma galectin‐3 levels (hazard ratio [HR] per 1 SD increase in natural log galectin‐3) and incident coronary heart disease, ischemic stroke, HF hospitalization, and total mortality in ARIC (Atherosclerosis Risk in Communities) participants free of cardiovascular disease at ARIC visit 4 (1996–1998; n=9247) and at ARIC visit 5 (2011–2013; n=4829). Higher galectin‐3 level at visit 4 (median age 62) was independently associated with incident coronary heart disease (adjusted HR, 1.30; 95% CI, 1.06–1.60), ischemic stroke (HR, 1.42; 95% CI, 1.01–2.00), HF (HR, 1.44; 95% CI, 1.17–1.76), and mortality (HR, 1.56; 95% CI, 1.35–1.80). At visit 5 (median age, 74), higher galectin‐3 level was associated with incident HF (HR, 1.93; 95% CI, 1.15–3.24) and total mortality (HR, 1.70; 95% CI, 1.15–2.52), but not coronary heart disease or stoke. Individuals with the greatest increase in galectin‐3 levels from visit 4 to visit 5 were also at increased risk of incident HF and total mortality. Conclusions In a large, biracial community‐based cohort, galectin‐3 measured at midlife and older age was associated with increased risk of cardiovascular events. An increase in galectin‐3 levels over this period was also associated with increased risk.
Collapse
Affiliation(s)
- David Aguilar
- Department of Epidemiology, Human Genetics, and Environmental Sciences School of Public Health University of Texas Health Science Center at Houston TX
| | | | - Ron C Hoogeveen
- Section of Cardiovascular Research Department of Medicine Baylor College of Medicine Houston TX.,Center for Cardiometabolic Disease Prevention Department of Medicine Baylor College of Medicine Houston TX
| | - Vijay Nambi
- Section of Cardiology Department of Medicine Baylor College of Medicine Houston TX.,Section of Cardiovascular Research Department of Medicine Baylor College of Medicine Houston TX.,Center for Cardiometabolic Disease Prevention Department of Medicine Baylor College of Medicine Houston TX.,Department of Medicine Michael E. DeBakey Veterans Affairs Medical Center Houston TX
| | - Elizabeth Selvin
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | - Kunihiro Matsushita
- Department of Epidemiology Johns Hopkins Bloomberg School of Public Health Baltimore MD
| | - Anum Saeed
- Section of Cardiovascular Research Department of Medicine Baylor College of Medicine Houston TX.,Heart and Vascular Institute University of Pittsburgh Medical Center Pittsburgh PA
| | - John W McEvoy
- Department of Medicine Ciccarone Center for the Prevention of Heart Disease Johns Hopkins School of Medicine Baltimore MD.,National University of Ireland and National Institute for Prevention and Cardiovascular Health Galway Ireland
| | - Amil M Shah
- Division of Cardiovascular Medicine Department of Medicine Brigham and Women's Hospital Boston MA
| | - Scott D Solomon
- Division of Cardiovascular Medicine Department of Medicine Brigham and Women's Hospital Boston MA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences School of Public Health University of Texas Health Science Center at Houston TX
| | - Christie M Ballantyne
- Section of Cardiology Department of Medicine Baylor College of Medicine Houston TX.,Section of Cardiovascular Research Department of Medicine Baylor College of Medicine Houston TX.,Center for Cardiometabolic Disease Prevention Department of Medicine Baylor College of Medicine Houston TX
| |
Collapse
|
154
|
Elevated galectin-3 in women with gestational diabetes mellitus, a new surrogate for cardiovascular disease in women. PLoS One 2020; 15:e0234732. [PMID: 32555606 PMCID: PMC7299386 DOI: 10.1371/journal.pone.0234732] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 06/01/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is associated with future cardiovascular morbidity and recognized as a women-specific risk factor for cardiovascular disease. The mechanisms for this association are not well established. Therefore, we aimed to evaluate the cardiovascular-related biomarkers, galectin-3 (Gal-3) and protein convertase subtilisin/kexin (PCSK) type 9, in women with GDM. METHODS Blood samples were drawn in the third trimester from 31 women diagnosed with GDM and from 35 women with normal pregnancies. Blood levels of Gal-3 and PCSK-9 were measured using a quantitative sandwich enzyme immunoassay. In addition, we measured Gal-3 levels in 24 pregnant women in the first trimester who later developed GDM and in 36 healthy controls. Continuous variables were compared using student's t-test and categorical variables by chi-square/fisher's exact tests. RESULTS We found increased levels of Gal-3 in women diagnosed with GDM compared to women without GDM (124.6±32% versus control; pv = 0.001). Furthermore, we demonstrated elevated levels of Gal-3 during the first trimester among women who later developed GDM compared with women who did not develop any gestational morbidity (125.7±32% versus control; pv = 0.004). Third-trimester levels of PCSK-9 did not differ between women with and without GDM (560±45ng/mL versus 553±33ng/mL; pv = 0.4). CONCLUSIONS The results suggest a possible mechanism that may link GDM to the future increased cardiovascular risk in these patients. Additionally, increased Gal-3 levels during the first trimester may suggest a new early predictor for GDM.
Collapse
|
155
|
Tian Y, Lv W, Lu C, Jiang Y, Yang X, Song M. Galectin-3 inhibition attenuates doxorubicin-induced cardiac dysfunction by upregulating the expression of peroxiredoxin-4. Can J Physiol Pharmacol 2020; 98:700-707. [PMID: 32516552 DOI: 10.1139/cjpp-2019-0700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Doxorubicin (DOX) is a highly efficient chemotherapeutic drug limited by its cardiotoxicity. Galectin-3 (Gal-3) overexpression is associated with several cardiovascular diseases. In this study, the in vivo models of DOX-treated rats and the in vitro model of DOX-treated H9C2 cells were used. DOX induced cardiac injury and dysfunction accompanied with the upregulation of Gal-3 at the end of the experiment, while inhibition of Gal-3 with modified citrus pectin (MCP) exhibited a dramatic improvement in cardiac function of the DOX-treated rats, as manifested by increased left ventricular systolic pressure and ±dp/dtmax and decreased left ventricular end-diastolic pressure. The plasma levels of myocardial injury markers such as lactate dehydrogenase, creatine kinase, creatine kinase-MB, and cardiac troponin I were decreased after MCP treatment. In parallel, MCP attenuated myocardial tissue markers of oxidative stress such as hydrogen peroxide and malondialdehyde restored the activities of superoxide dismutase, catalase, and glutathione peroxidase and upregulated antioxidant peroxiredoxin-4 (Prx-4). To further verify the role of Prx-4, it was downregulated by siRNA-mediated knockdown in H9C2 cells. MCP could not reverse DOX-induced oxidative stress in Prx-4-knock-down cells. In conclusion, Gal-3 mediated DOX-induced cardiotoxicity and Gal-3 inhibition attenuated DOX-induced cardiac dysfunction by upregulating the expression of Prx-4 to reduce myocardial oxidative stress.
Collapse
Affiliation(s)
- Yunpeng Tian
- Department of Cardiology and Department of Cardiac Surgery, Tianjin First Central Hospital, Tianjin 300192, People's Republic of China
| | - Wei Lv
- Department of Cardiology and Department of Cardiac Surgery, Tianjin First Central Hospital, Tianjin 300192, People's Republic of China
| | - Chengzhi Lu
- Department of Cardiology and Department of Cardiac Surgery, Tianjin First Central Hospital, Tianjin 300192, People's Republic of China
| | - Yiyao Jiang
- Department of Cardiology and Department of Cardiac Surgery, Tianjin First Central Hospital, Tianjin 300192, People's Republic of China
| | - Xue Yang
- Department of Cardiology, Dalian Municipal Central Hospital, Dalian 116003, People's Republic of China
| | - Minghao Song
- Department of Cardiology, Tongji Hospital of Tongji University, Shanghai 200003, People's Republic of China
| |
Collapse
|
156
|
Wu D, Zheng J, Hu W, Zheng X, He Q, Linhardt RJ, Ye X, Chen S. Structure-activity relationship of Citrus segment membrane RG-I pectin against Galectin-3: The galactan is not the only important factor. Carbohydr Polym 2020; 245:116526. [PMID: 32718630 DOI: 10.1016/j.carbpol.2020.116526] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 11/25/2022]
Abstract
Rhamnogalacturonan I (RG-I) pectin are regarded as strong galectin-3 (Gal-3) antagonist because of galactan sidechains. The present study focused on discussing the effects of more structural regions in pectin on the anti-Gal-3 activity. The water-soluble pectin (WSP) recovered from citrus canning processing water was categorized as RG-I pectin. The controlled enzymatic hydrolysis was employed to sequentially remove the α-1,5-arabinan, homogalaturonan and β-1,4-galactan in WSP. The Gal-3-binding affinity KD (kd/ka) of WSP and debranched pectins were calculated to be 0.32 μM, 0.48 μM, 0.56 μM and 1.93 μM. Moreover, based on the more sensitive cell line (MCF-7) model, the IC30 value of WSP was lower than these of modified pectins, indicating decreased anti-Gal-3 activity. Our results suggested that the total amount of neutral sugar sidechains, the length of arabinan and cooperation between HG and RG-I played important roles in the anti-Gal-3 activity of WSP, not the Gal/Ara ratio or RG-I/HG ratio. These results provided a new insight into structure-activity relationship of citrus segment membrane RG-I as a galectin-3 antagonist and a new functional food.
Collapse
Affiliation(s)
- Dongmei Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Jiaqi Zheng
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Weiwei Hu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China
| | - Xiaoliang Zheng
- Center for Molecular Medicine, Zhejiang Academy of Medical Sciences, Hangzhou 310013, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Integrated Research Base of Southern Fruit and Vegetable Preservation Technology, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou 310058, China; Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China.
| |
Collapse
|
157
|
Berezin AE, Berezin AA. Circulating Cardiac Biomarkers in Diabetes Mellitus: A New Dawn for Risk Stratification-A Narrative Review. Diabetes Ther 2020; 11:1271-1291. [PMID: 32430864 PMCID: PMC7261294 DOI: 10.1007/s13300-020-00835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
The aim of this narrative review is to update the current knowledge on the differential choice of circulating cardiac biomarkers in patients with prediabetes and established type 2 diabetes mellitus (T2DM). There are numerous circulating biomarkers with unconfirmed abilities to predict clinical outcomes in pre-DM and DM individuals; the prognostication ability of the cardiac biomarkers reported here has been established, and they are still being studied. The conventional cardiac biomarkers, such as natriuretic peptides (NPs), soluble suppressor tumorigenisity-2, high-sensitivity circulating cardiac troponins and galectin-3, were useful to ascertain cardiovascular (CV) risk. Each cardiac biomarker has its strengths and weaknesses that affect the price of usage, specificity, sensitivity, predictive value and superiority in face-to-face comparisons. Additionally, there have been confusing reports regarding their abilities to be predictably relevant among patients without known CV disease. The large spectrum of promising cardiac biomarkers (growth/differential factor-15, heart-type fatty acid-binding protein, cardiotrophin-1, carboxy-terminal telopeptide of collagen type 1, apelin and non-coding RNAs) is discussed in the context of predicting CV diseases and events in patients with known prediabetes and T2DM. Various reasons have been critically discussed related to the variable findings regarding biomarker-based prediction of CV risk among patients with metabolic disease. It was found that NPs and hs-cTnT are still the most important tools that have an affordable price as well as high sensitivity and specificity to predict clinical outcomes among patients with pre-DM and DM in routine clinical practice, but other circulating biomarkers need to be carefully investigated in large trials in the future.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, Ministry of Health of Ukraine, State Medical University, Zaporozhye, 69035, Ukraine.
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, 69096, Ukraine
| |
Collapse
|
158
|
Cohen MD, Prophete C, Horton L, Sisco M, Park SH, Lee HW, Zelikoff J, Chen LC. Impact on rats from acute intratracheal inhalation exposures to WTC dusts. Inhal Toxicol 2020; 32:218-230. [PMID: 32448006 DOI: 10.1080/08958378.2020.1768322] [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] [Indexed: 10/24/2022]
Abstract
Background: Studies have revealed the increased incidence of health disorders in First Responders (FR) who were at Ground Zero over the initial 72 hr after the World Trade Center (WTC) collapses. Previous studies in rats exposed to WTC dusts using exposure scenarios that mimicked FR mouthbreathing showed exposure led to altered expression of genes whose products could be involved in lung ailments. Nevertheless, it was uncertain if repeated exposures (as occurred in earliest days post-disaster) might have given rise to long-term changes in the lungs/other organs, in white blood cell (WBC) profiles, and/or systemic expression of select (mostly immune-related) proteins.Methods: To examine this, rats were exposed on 2 consecutive days (2 hr/d, intratracheal inhalation) to WTC dusts and then examined over a 1-yr period thereafter. At select times post-exposure, organ (lung, heart, liver, kidney, spleen) weights, WBC profiles, and blood levels of a variety of proteins were evaluated.Results: The study showed that over the 1-yr period, there were nominal effects on organ weights (absolute, index) as a result of the dust exposures. There were significant changes (relative to in naïve rats) in WBC profiles, with exposed rats having increased monocyte-macrophage and decreased lymphocyte percentages. The study also found that dust exposure led to significant systemic increases in many proteins, including MCP-1, RANTES, MMP-9, RAGE, and Galectin-3.Conclusions: These results provide further support for our longstanding hypothesis that the WTC dusts could potentially have acted as direct inducers of many of the health effects that have been seen in the exposed FR.
Collapse
Affiliation(s)
- Mitchell D Cohen
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Colette Prophete
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Lori Horton
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Maureen Sisco
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Sung-Hyun Park
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Hyun-Wook Lee
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Judith Zelikoff
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| | - Lung-Chi Chen
- Department of Environmental Medicine, New York University of School of Medicine, New York, NY, USA
| |
Collapse
|
159
|
Song B, Yao B, Dang H, Dong R. Soluble ST2, Galectin-3 and clinical prognosis of patients with hypertrophic cardiomyopathy undergoing ventricular septal myectomy: a correlation analysis. Cardiovasc Diagn Ther 2020; 10:145-152. [PMID: 32420094 DOI: 10.21037/cdt.2020.01.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is the most common chromosomal abnormal heart disease. The pathophysiological mechanism of HCM is complex. Several studies have suggested that the level of Soluble ST2 (sST2) may be a biomarker of chronic systolic heart failure, however, the role of sST2 in HCM remains unclear. So we performed this study to analyze the role of Soluble ST2 (sST2), Galectin-3 (Gal-3) and its correlations with clinical prognosis of patients with hypertrophic cardiomyopathy (HCM) undergoing ventricular septal myectomy. Methods HCM patients who underwent modified Morrow surgery in our hospital during June 2016-June 2018 were included. We divided the patients into different groups stratified by sST2 and Gal-3 level. Besides, we included volunteers without heart disease for medical examination as normal controls. Biochemical analyses were conducted to identify the biomarkers difference. The predictive value of sST2 and Gal-3 on all-cause mortality was evaluated with Cox regression analysis. Results A total of 125 HCM patients were included in this present study. The sST2 and Gal-3 levels in HCM patients were significantly higher than that in control group (all P<0.001); there were significant differences in the incidence of all-cause mortality for HCM patients stratified by the sST2 and Gal-3 level; Cox univariate regression survival analysis showed that the hypertension (HR =1.19, 95% CI: 1.01-1.38), maximum wall thickness (HR =1.48, 95% CI: 1.04-1.98), Log sST2 (HR =1.02, 95% CI: 1.01-1.05), Log Gal-3 (HR =1.17, 95% CI: 1.09-1.32) were the predictors for all-cause mortality in patients with HCM, and Cox multivariate risk regression showed that maximum wall thickness was the independent predictors of all-cause mortality in patients with HCM (HR =1.63, 95% CI: 1.35-1.97). Conclusions Even through sST2 and Gal-3 were not associated with clinical prognosis of patients with HCM undergoing ventricular septal myectomy, it may be involved in the progress of HCM, more studies are warranted to identify the potential mechanism and reverence value.
Collapse
Affiliation(s)
- Bangrong Song
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Bo Yao
- Cardiology Section 2, Zibo City Linzi District People's Hospital, Zibo 255400, China
| | - Haiming Dang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ran Dong
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
160
|
Plasma Galectin-3 predicts deleterious vascular dysfunction affecting post-myocardial infarction patients: An explanatory study. PLoS One 2020; 15:e0232572. [PMID: 32392260 PMCID: PMC7213735 DOI: 10.1371/journal.pone.0232572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 04/16/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES In a previous analysis of a post-myocardial infarction (MI) cohort, abnormally high systemic vascular resistances (SVR) were shown to be frequently revealed by MRI during the healing period, independently of MI severity, giving evidence of vascular dysfunction and limiting further recovery of cardiac function. The present ancillary and exploratory analysis of the same cohort was aimed at characterizing those patients suffering from high SVR remotely from MI with a large a panel of cardiovascular MRI parameters and blood biomarkers. METHODS MRI and blood sampling were performed 2-4 days after a reperfused MI and 6 months thereafter in 121 patients. SVR were monitored with a phase-contrast MRI sequence and patients with abnormally high SVR at 6-months were characterized through MRI parameters and blood biomarkers, including Galectin-3, an indicator of cardiovascular inflammation and fibrosis after MI. SVR were normal at 6-months in 90 patients (SVR-) and abnormally high in 31 among whom 21 already had high SVR at the acute phase (SVR++) while 10 did not (SVR+). RESULTS When compared with SVR-, both SVR+ and SVR++ exhibited lower recovery in cardiac function from baseline to 6-months, while baseline levels of Galectin-3 were significantly different in both SVR+ (median: 14.4 (interquartile range: 12.3-16.7) ng.mL-1) and SVR++ (13.0 (11.7-19.4) ng.mL-1) compared to SVR- (11.7 (9.8-13.5) ng.mL-1, both p < 0.05). Plasma Galectin-3 was an independent baseline predictor of high SVR at 6-months (p = 0.002), together with the baseline levels of SVR and left ventricular end-diastolic volume, whereas indices of MI severity and left ventricular function were not. In conclusion, plasma Galectin-3 predicts a deleterious vascular dysfunction affecting post-MI patients, an observation that could lead to consider new therapeutic targets if confirmed through dedicated prospective studies.
Collapse
|
161
|
Suthahar N, Meems LMG, Ho JE, de Boer RA. Sex-related differences in contemporary biomarkers for heart failure: a review. Eur J Heart Fail 2020; 22:775-788. [PMID: 32220046 PMCID: PMC7319414 DOI: 10.1002/ejhf.1771] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/28/2022] Open
Abstract
The use of circulating biomarkers for heart failure (HF) is engrained in contemporary cardiovascular practice and provides objective information about various pathophysiological pathways associated with HF syndrome. However, biomarker profiles differ considerably among women and men. For instance, in the general population, markers of cardiac stretch (natriuretic peptides) and fibrosis (galectin‐3) are higher in women, whereas markers of cardiac injury (cardiac troponins) and inflammation (sST2) are higher in men. Such differences may reflect sex‐specific pathogenic processes associated with HF risk, but may also arise as a result of differences in sex hormone profiles and fat distribution. From a clinical perspective, sex‐related differences in biomarker levels may affect the objectivity of biomarkers in HF management because what is considered to be ‘normal’ in one sex may not be so in the other. The objectives of this review are, therefore: (i) to examine the sex‐specific dynamics of clinically relevant HF biomarkers in the general population, as well as in HF patients; (ii) to discuss the overlap between sex‐related and obesity‐related effects, and (iii) to identify knowledge gaps to stimulate research on sex‐related differences in
HF.
Collapse
Affiliation(s)
- Navin Suthahar
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Laura M G Meems
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Jennifer E Ho
- Division of Cardiology, Department of Medicine, and Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rudolf A de Boer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| |
Collapse
|
162
|
Abstract
Cardiac fibrosis is associated with non-ischemic dilated cardiomyopathy, increasing its morbidity and mortality. Cardiac fibroblast is the keystone of fibrogenesis, being activated by numerous cellular and humoral factors. Macrophages, CD4+ and CD8+ T cells, mast cells, and endothelial cells stimulate fibrogenesis directly by activating cardiac fibroblasts and indirectly by synthetizing various profibrotic molecules. The synthesis of type 1 and type 3 collagen, fibronectin, and α-smooth muscle actin is rendered by various mechanisms like transforming growth factor-beta/small mothers against decapentaplegic pathway, renin angiotensin system, and estrogens, which in turn alter the extracellular matrix. Investigating the underlying mechanisms will allow the development of diagnostic and prognostic tools and discover novel specific therapies. Serum biomarkers aid in the diagnosis and tracking of cardiac fibrosis progression. The diagnostic gold standard is cardiac magnetic resonance with gadolinium administration that allows quantification of cardiac fibrosis either by late gadolinium enhancement assessment or by T1 mapping. Therefore, the goal is to stop and even reverse cardiac fibrosis by developing specific therapies that directly target fibrogenesis, in addition to the drugs used to treat heart failure. Cardiac resynchronization therapy had shown to revert myocardial remodeling and to reduce cardiac fibrosis. The purpose of this review is to provide an overview of currently available data.
Collapse
|
163
|
Coats AJS. Figures of the Heart Failure Association (HFA): Dr. Rudolf de Boer, HFA Board Member (2014-2020), Chair of the Basic Science Section (2016-2018), coordinator of the Study Group on Heart Failure with Preserved Ejection Fraction, and member of the HFA study groups of Translational Research and Cardio-oncology. Eur J Heart Fail 2020; 22:572-575. [PMID: 32128968 DOI: 10.1002/ejhf.1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 11/09/2022] Open
|
164
|
Kasacka I, Piotrowska Ż, Niezgoda M, Lewandowska A, Łebkowski W. Ageing-related changes in the levels of β-catenin, CacyBP/SIP, galectin-3 and immunoproteasome subunit LMP7 in the heart of men. PLoS One 2020; 15:e0229462. [PMID: 32119722 PMCID: PMC7051089 DOI: 10.1371/journal.pone.0229462] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 02/06/2020] [Indexed: 01/12/2023] Open
Abstract
Aging is a major risk factor for morbidity and mortality from cardiovascular causes in men. To better understand the cellular processes related to age-related cardiac complications, we undertook research aimed at comparative evaluation of genes expression and distribution of β-catenin, CacyBP/SIP, galectin-3 and LMP7 in the heart of healthy men in different age groups. The study was conducted on the hearts of 12 men (organ donors) without a history of cardiovascular disease, who were divided into two age groups: men under and men over 45 years of age. On paraffin sections, immunohistochemical reactions were performed to detect β-catenin, CacyBP/SIP, galectin-3 and immunoproteasome subunit LMP7. The expression of genes coding β-catenin, CacyBP/SIP, galectin-3 and LMP7 was also evaluated by real-time PCR method. In the heart of men over 45 years old, both gene expression and immunoreactivity of β-catenin, CacyBP/SIP, galectin-3 and LMP7 were stronger compared to younger individuals. The results of the presented studies suggest that β-catenin, CacyBP/SIP, galectin-3 and immunoproteasomes might be involved in the internal regulation of heart homeostasis during ageing.
Collapse
Affiliation(s)
- Irena Kasacka
- Department of Histology and Cytophysiology, Medical University of Białystok, Białystok, Poland
- * E-mail: ,
| | - Żaneta Piotrowska
- Department of Histology and Cytophysiology, Medical University of Białystok, Białystok, Poland
| | - Michał Niezgoda
- Department of Histology and Cytophysiology, Medical University of Białystok, Białystok, Poland
| | - Alicja Lewandowska
- Department of Histology and Cytophysiology, Medical University of Białystok, Białystok, Poland
| | - Wojciech Łebkowski
- Department of Neurosurgery, Medical University of Bialystok, Białystok, Poland
| |
Collapse
|
165
|
Chen H, Chen C, Fang J, Wang R, Nie W. Circulating galectin-3 on admission and prognosis in acute heart failure patients: a meta-analysis. Heart Fail Rev 2020; 25:331-341. [PMID: 31641977 DOI: 10.1007/s10741-019-09858-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Changes of serum galectin-3 have been associated with the pathogenesis of many cardiovascular diseases. The aim of the study was to evaluate the prognostic role of serum galectin-3 in patients with acute heart failure (AHF) in a meta-analysis. Follow-up studies evaluating the association between serum galectin-3 on admission and clinical outcomes in AHF patients were identified via search of PubMed and Embase databases. A random effects or a fixed effects model was applied to pool the results depending on the heterogeneity. Subgroup analysis was used to evaluate the influences of study characteristics on the outcomes. Overall, 7057 AHF patients from eighteen follow-up studies were included. Higher serum galectin-3 was associated with higher risks of all-cause mortality (adjusted risk ratio [RR], 1.58; p < 0.001), mortality/HF rehospitalization (RR, 1.68; p < 0.001), and cardiovascular mortality (RR, 1.29; p = 0.04), but not HF rehospitalization (RR, 1.24; p = 0.25) in AHF patients. Subgroup analyses showed that study characteristics including study design, sample size, age, gender, left ventricular ejection fraction, galectin-3 variable type, follow-up duration, and adjustment of type B natriuretic peptide did not significantly impact the results. Significant heterogeneities were detected for the outcomes of all-cause mortality and mortality/HF rehospitalization. However, trim-and-fill analyses by including the imputed studies to generate symmetrical funnel plots showed similar significant meta-analysis results. These results suggested that higher serum galectin-3 may be associated with poor prognosis in AHF patients. Further studies are needed to determine the mechanisms underlying the potential prognostic role of galectin-3 in AHF.
Collapse
Affiliation(s)
- Hongsen Chen
- Department of Intensive Care Unit, The First People's Hospital of Xiangshan, No. 291 Donggu Road, Xiangshan County, Ningbo, 315700, China.
| | - Chensong Chen
- Department of Intensive Care Unit, The First People's Hospital of Xiangshan, No. 291 Donggu Road, Xiangshan County, Ningbo, 315700, China
| | - Junjie Fang
- Department of Intensive Care Unit, The First People's Hospital of Xiangshan, No. 291 Donggu Road, Xiangshan County, Ningbo, 315700, China
| | - Ren Wang
- Department of Intensive Care Unit, The First People's Hospital of Xiangshan, No. 291 Donggu Road, Xiangshan County, Ningbo, 315700, China
| | - Wanshui Nie
- Department of Intensive Care Unit, The First People's Hospital of Xiangshan, No. 291 Donggu Road, Xiangshan County, Ningbo, 315700, China
| |
Collapse
|
166
|
Galectin-3 Is a Potential Mediator for Atherosclerosis. J Immunol Res 2020; 2020:5284728. [PMID: 32149158 PMCID: PMC7042544 DOI: 10.1155/2020/5284728] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/13/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic inflammatory arterial disease forming the pathological basis of many cardiovascular diseases such as coronary heart disease, heart failure, and stroke. Numerous studies have implicated inflammation as a key player in the initiation and progression of atherosclerosis. Galectin-3 (Gal-3) is a 30 kDa β-galactose, highly conserved and widely distributed intracellularly and extracellularly. Gal-3 has been demonstrated in recent years to be a novel inflammatory factor participating in the process of intravascular inflammation, lipid endocytosis, macrophage activation, cellular proliferation, monocyte chemotaxis, and cell adhesion. This review focuses on the role of Gal-3 in atherosclerosis and the mechanism involved and several classical Gal-3 agonists and antagonists in the current studies.
Collapse
|
167
|
Patel DM, Thiessen-Philbrook H, Brown JR, McArthur E, Moledina DG, Mansour SG, Shlipak MG, Koyner JL, Kavsak P, Whitlock RP, Everett AD, Malenka DJ, Garg AX, Coca SG, Parikh CR. Association of plasma-soluble ST2 and galectin-3 with cardiovascular events and mortality following cardiac surgery. Am Heart J 2020; 220:253-263. [PMID: 31911262 PMCID: PMC7008086 DOI: 10.1016/j.ahj.2019.11.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 11/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Cardiac surgery induces hemodynamic stress on the myocardium, and this process can be associated with significant post-operative morbidity and mortality. Soluble suppression of tumorigenicity 2 (sST2) and galectin-3 (gal-3) are biomarkers of myocardial remodeling and fibrosis; however, their potential association with post-operative changes is unknown. METHODS We measured peri-operative plasma sST2 and gal-3 levels in two prospective cohorts (TRIBE-AKI and NNE) of over 1800 patients who underwent cardiac surgery. sST2 and gal-3 levels were evaluated for association with a composite primary outcome of cardiovascular event or mortality over median follow-up periods of 3.4 and 6.0 years, respectively, for the two cohorts. Meta-analysis of hazard ratio estimates from the cohorts was performed using random effects models. RESULTS Cohorts demonstrated event rates of 70.2 and 66.8 per 1000 person-years for the primary composite outcome. After adjustment for clinical covariates, higher post-operative sST2 and gal-3 levels were significantly associated with cardiovascular event or mortality [pooled estimate HRs: sST2 1.29 (95% CI 1.16, 1.44); gal-3 1.26 (95% CI 1.09, 1.46)]. These associations were not significantly modified by pre-operative congestive heart failure or AKI. CONCLUSIONS Higher post-operative sST2 and gal-3 values were associated with increased incidence of cardiovascular event or mortality. These two biomarkers should be further studied for potential clinical utility for patients undergoing cardiac surgery.
Collapse
Affiliation(s)
- Dipal M Patel
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT
| | | | - Jeremiah R Brown
- Dartmouth Institute for Health Policy and Clinical Practice, and the Departments of Biomedical Data Science and Epidemiology, Geisel School of Medicine, Lebanon, NH
| | | | - Dennis G Moledina
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Sherry G Mansour
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Michael G Shlipak
- Kidney Health Research Collaborative, University of California San Francisco, San Francisco, CA; Department of Medicine, San Francisco VA Medical Center and University of California, San Francisco, CA
| | - Jay L Koyner
- Section of Nephrology, Department of Medicine, University of Chicago, Pritzker School of Medicine, Chicago, IL
| | - Peter Kavsak
- Departments of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Richard P Whitlock
- Population Health Research Institute and Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Allen D Everett
- Division of Cardiology, Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - David J Malenka
- Dartmouth Institute for Health Policy and Clinical Practice, and the Departments of Biomedical Data Science and Epidemiology, Geisel School of Medicine, Lebanon, NH
| | - Amit X Garg
- ICES, Toronto, ON, Canada; Division of Nephrology, Department of Medicine, Western University, London, ON, Canada
| | - Steven G Coca
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Chirag R Parikh
- Division of Nephrology, School of Medicine, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
168
|
|
169
|
Roles of galectin-3 in metabolic disorders and tumor cell metabolism. Int J Biol Macromol 2020; 142:463-473. [DOI: 10.1016/j.ijbiomac.2019.09.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/03/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
|
170
|
Favorable Response to CD34+ Cell Therapy Is Associated with a Decrease of Galectin-3 Levels in Patients with Chronic Heart Failure. DISEASE MARKERS 2019; 2019:8636930. [PMID: 31885743 PMCID: PMC6925830 DOI: 10.1155/2019/8636930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
Background Galectin-3 plasma levels (gal-3) were shown to correlate with the scar burden in chronic heart failure (CHF) setting. As scar burden predicts response to stem cell therapy, we sought to explore a correlation between gal-3 and response to CD34+ cell transplantation in patients with CHF. Methods We performed a post hoc analysis of patients, enrolled in 2 prospective trials investigating the clinical effects of CD34+ cell therapy in patients with ischemic cardiomyopathy (ICMP) and nonischemic dilated cardiomyopathy (DCMP). CD34+ cells were mobilized by G-CSF, collected via apheresis, and injected transendocardially using NOGA system. Patients were followed for 3 months and demographic, echocardiographic, and biochemical parameters and gal-3 were analyzed at baseline and at follow-up. Response to cell therapy was defined as an LVEF increase of ≥5%. Results 61 patients were included in the analysis. The mean age of patients was 52 years and 83% were male. DCMP and ICMP were present in 69% and 31% of patients, respectively. The average serum creatinine was 86 ± 23 μmol/L, NT-proBNP 1132 (IQR 350-2279) pg/mL, and LVEF 30 ± 6%. Gal-3 at baseline and at 3 months did not differ significantly (13.4 ± 5.5 ng/mL vs. 13.1 ± 5.8 ng/mL; p = 0.72), and there were no differences in baseline gal-3 with respect to heart failure etiology (15.1 ± 7.2 ng/mL in ICMP vs. 12.7 ± 4.3 ng/mL in DCMP; p = 0.12). Comparing responders (N = 49) to nonresponders (N = 18), we found no differences in baseline gal-3 (13.6 ± 5.7 ng/mL vs. 13.2 ± 4.9 ng/mL; p = 0.80). However, responders had significantly lower gal-3 at 3-month follow-up (12.1 ± 4.0 ng/mL vs. 15.7 ± 8.4 ng/mL; p < 0.05). Also, responders demonstrated a significant decrease in gal-3 over 3 months, while in nonresponders, an increase in gal-3 occurred (−1.5 ± 5.4 ng/mL vs. +2.7 ± 4.3 ng/mL; p = 0.01). Conclusions In patients with chronic heart failure undergoing CD34+ cell therapy, a decrease in galectin-3 plasma levels is associated with beneficial response to this treatment modality. Further prospective data is warranted to confirm our findings and to deepen our understanding of the role of gal-3 in the field of stem cell therapy.
Collapse
|
171
|
Dhir S, Dhir A. Cardiovascular Risk Assessment for Noncardiac Surgery: Are We Ready for Biomarkers? J Cardiothorac Vasc Anesth 2019; 34:1914-1924. [PMID: 31866221 DOI: 10.1053/j.jvca.2019.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 09/07/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023]
Abstract
Biomarkers aided perioperative cardiac assessment is a relatively new concept. Cardiac biomarkers with historical significance (aspartate transaminase, dehydrogenase, creatinine kinase and myoglobin) have paved the way for traditional biomarkers (cardiac troponin, C-reactive protein, lipoprotein). Contemporary biomarkers like natriuretic peptides (BNP and ProBNP) are validated risk markers in both acute and chronic cardiac diseases and are showing remarkable promise in predicting serious cardiovascular complications after non-cardiac surgery. This review is intended to provide a critical overview of traditional and contemporary biomarkers for perioperative cardiovascular assessment and management. This review also discusses the potential utility of newer biomarkers like galectin-3, sST-2, GDF-15, TNF-alpha, MiRNAs and many others that can predict inflammation, cardiac remodeling, injury and endogenous stress and need further investigations to establish their clinical utility. Though promising, biomarker led perioperative care is still in infancy and it has not been determined that it can improve clinical outcomes.
Collapse
Affiliation(s)
- Shalini Dhir
- Department of Anesthesia and Perioperative Medicine, Western University, London, Ontario, Canada.
| | - Achal Dhir
- Department of Anesthesia and Perioperative Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
172
|
Zhang X, Karunathilaka N, Senanayake S, Subramaniam VN, Chan W, Kostner K, Fraser J, Atherton JJ, Punyadeera C. The potential prognostic utility of salivary galectin-3 concentrations in heart failure. Clin Res Cardiol 2019; 109:685-692. [PMID: 31598750 DOI: 10.1007/s00392-019-01557-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/24/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Patients with HF are at a higher risk of rehospitalisation and, as such, significant costs to our healthcare system. A non-invasive method to collect body fluids and measure Gal-3 could improve the current management of HF. In this study, we investigated the potential prognostic utility of salivary Galectin-3 (Gal-3) in patients with heart failure (HF). METHODS We collected saliva samples from patients with HF (n = 105) either at hospital discharge or during routine clinical visits. Gal-3 concentrations in saliva samples were measured by ELISA. The Kaplan-Meier survival curve analysis and Cox proportional regression model were used to determine the potential prognostic utility of salivary Gal-3 concentrations. RESULTS The primary end point was either cardiovascular death or hospitalisation. Salivary Gal-3 concentrations were significantly higher (p < 0.05) in patients with HF who subsequently experienced the primary endpoint compared to those who did not. HF patients with salivary Gal-3 concentrations > 172.58 ng/mL had a significantly (p < 0.05) higher cumulative risk of the primary endpoint compared to those with lower salivary Gal-3 concentrations. In patients with HF, salivary Gal-3 concentration was a predictor of the primary endpoint even after adjusting for other covariates. CONCLUSIONS In our pilot study, HF patients with salivary Gal-3 concentrations of > 172.58 ng/mL demonstrated a higher cumulative risk of the primary outcome compared to those with lower Gal-3 levels, even after adjusting for other variables. Confirming our findings in a larger multi-centre clinical trial in the future would enable salivary Gal-3 measurements to form part of routine management for patients with HF.
Collapse
Affiliation(s)
- Xi Zhang
- Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, GPO Box 2434, Brisbane, QLD, 4001, Australia
| | - Nuwan Karunathilaka
- Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, GPO Box 2434, Brisbane, QLD, 4001, Australia
| | - Sameera Senanayake
- Australian Centre For Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - V Nathan Subramaniam
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Wandy Chan
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Karam Kostner
- Department of Cardiology, Mater Adult Hospital, Brisbane, QLD, Australia
| | - John Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - John J Atherton
- Cardiology Department, Royal Brisbane and Women's Hospital and University of Queensland School of Medicine, Brisbane, QLD, Australia
| | - Chamindie Punyadeera
- Saliva and Liquid Biopsy Translational Research Team, School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, GPO Box 2434, Brisbane, QLD, 4001, Australia.
| |
Collapse
|
173
|
Mo D, Tian W, Zhang HN, Feng YD, Sun Y, Quan W, Hao XW, Wang XY, Liu XX, Li C, Cao W, Liu WJ, Li XQ. Cardioprotective effects of galectin-3 inhibition against ischemia/reperfusion injury. Eur J Pharmacol 2019; 863:172701. [PMID: 31568784 DOI: 10.1016/j.ejphar.2019.172701] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023]
Abstract
Myocardial ischemia/reperfusion (IR) injury is caused by the restoration of the coronary blood flow following an ischemic episode. Accumulating evidence suggests that galectin-3, a β-galactoside-binding lectin, acts as a biomarker in heart disease. However, it remains unclear whether manipulating galectin-3 affects the susceptibility of the heart to IR injury. In this study, RNA sequencing (RNA-seq) analysis identified that Lgals3 (galecin-3) plays an indispensable role in IR-induced cardiac damage. Immunostaining and immunoblot assays confirmed that the expression of galectin-3 was markedly increased in myocardial IR injury both in vivo and in vitro. Echocardiographic analysis showed that cardiac dysfunction in experimental IR injury was significantly attenuated by galectin-3 inhibitors including pectin (1%, i.p.) from citrus and binding peptide G3-C12 (5.0 mg/kg, i.p.). Galectin-3 inhibitor-treated mice exhibited smaller infarct sizes and decreased tissue injury. Furthermore, TUNEL staining showed that galectin-3 inhibition suppressed IR-mediated cardiomyocyte apoptosis. Mitochondrial membrane potential (MMP) and mitochondrial permeability transition pore (mPTP) levels were well-preserved and IR-induced changes of mitochondrial cyto c, cytosol cyto c, caspase-9, caspase-3, Bcl-2 and Bax in the galectin-3 inhibitor-treated groups were observed. Our findings indicate that the pathological upregulation of galectin-3 contributes to IR-induced cardiac dysfunction and that galectin-3 inhibition ameliorates myocardial injury, highlighting its therapeutic potential.
Collapse
Affiliation(s)
- Dan Mo
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Wen Tian
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Hui-Nan Zhang
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ying-Da Feng
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yang Sun
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Wei Quan
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiao-Wei Hao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xue-Ying Wang
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xiao-Xiao Liu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Chen Li
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Wei Cao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Wen-Juan Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, China
| | - Xiao-Qiang Li
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
174
|
Bogaard HJ, Voelkel NF. Is Myocardial Fibrosis Impairing Right Heart Function? Am J Respir Crit Care Med 2019; 199:1458-1459. [PMID: 30608865 PMCID: PMC6580680 DOI: 10.1164/rccm.201812-2307ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
175
|
Du X, Zhao W, Nguyen M, Lu Q, Kiriazis H. β-Adrenoceptor activation affects galectin-3 as a biomarker and therapeutic target in heart disease. Br J Pharmacol 2019; 176:2449-2464. [PMID: 30756388 PMCID: PMC6592856 DOI: 10.1111/bph.14620] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/11/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a key histopathological component that drives the progression of heart disease leading to heart failure and constitutes a therapeutic target. Recent preclinical and clinical studies have implicated galectin-3 (Gal-3) as a pro-fibrotic molecule and a biomarker of heart disease and fibrosis. However, our knowledge is poor on the mechanism(s) that determine the blood level or regulate cardiac expression of Gal-3. Recent studies have demonstrated that enhanced β-adrenoceptor activity is a determinant of both circulating concentration and cardiac expression of Gal-3. Pharmacological or transgenic activation of β-adrenoceptors leads to increased blood levels of Gal-3 and up-regulated cardiac Gal-3 expression, effect that can be reversed with the use of β-adrenoceptor antagonists. Conversely, Gal-3 gene deletion confers protection against isoprenaline-induced cardiotoxicity and fibrogenesis. At the transcription level, β-adrenoceptor stimulation activates cardiac mammalian sterile-20-like kinase 1, a pivotal kinase of the Hippo signalling pathway, which is associated with Gal-3 up-regulation. Recent studies have suggested a role for the β-adrenoceptor-Hippo signalling pathway in the regulation of cardiac Gal-3 expression thereby contributing to the onset and progression of heart disease. This implies a therapeutic potential of the suppression of Gal-3 expression. In this review, we discuss the effects of β-adrenoceptor activity on Gal-3 as a biomarker and causative mediator in the setting of heart disease and point out pivotal knowledge gaps. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Xiao‐Jun Du
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Physiology and Pathophysiology, School of Medical SciencesXi'an Jiaotong University Health Science CenterXi'anChina
| | - Wei‐Bo Zhao
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - My‐Nhan Nguyen
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| | - Qun Lu
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
- Department of Cardiovascular Medicine, First HospitalXi'an Jiaotong University Health Science CenterXi'anChina
| | - Helen Kiriazis
- Experimental Cardiology LaboratoryBaker Heart and Diabetes InstituteMelbourneVICAustralia
| |
Collapse
|
176
|
Zhao WB, Lu Q, Nguyen MN, Su Y, Ziemann M, Wang LN, Kiriazis H, Puthalakath H, Sadoshima J, Hu HY, Du XJ. Stimulation of β-adrenoceptors up-regulates cardiac expression of galectin-3 and BIM through the Hippo signalling pathway. Br J Pharmacol 2019; 176:2465-2481. [PMID: 30932177 PMCID: PMC6592853 DOI: 10.1111/bph.14674] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/29/2019] [Accepted: 03/04/2019] [Indexed: 01/01/2023] Open
Abstract
Background and Purpose Expression of the pro‐fibrotic galectin‐3 and the pro‐apoptotic BIM is elevated in diseased heart or after β‐adrenoceptor stimulation, but the underlying mechanisms are unclear. This question was addressed in the present study. Experimental Approach Wild‐type mice and mice with cardiac transgenic expression of β2‐adrenoceptors, mammalian sterile‐20 like kinase 1 (Mst1) or dominant‐negative Mst1, and non‐specific galectin‐3 knockout mice were used. Effects of the β‐adrenoceptor agonist isoprenaline or β‐adrenoceptor antagonists were studied. Rat cardiomyoblasts (H9c2) were used for mechanistic exploration. Biochemical assays were performed. Key Results Isoprenaline treatment up‐regulated expression of galectin‐3 and BIM, and this was inhibited by non‐selective or selective β‐adrenoceptor antagonists (by 60–70%). Cardiac expression of galectin‐3 and BIM was increased in β2‐adrenoceptor transgenic mice. Isoprenaline‐induced up‐regulation of galectin‐3 and BIM was attenuated by Mst1 inactivation, but isoprenaline‐induced galectin‐3 expression was exaggerated by transgenic Mst1 activation. Pharmacological or genetic activation of β‐adrenoceptors induced Mst1 expression and yes‐associated protein (YAP) phosphorylation. YAP hyper‐phosphorylation was also evident in Mst1 transgenic hearts with up‐regulated expression of galectin‐3 (40‐fold) and BIM as well as up‐regulation of many YAP‐target genes by RNA sequencing. In H9c2 cells, isoprenaline induced YAP phosphorylation and expression of galectin‐3 and BIM, effects simulated by forskolin but abolished by PKA inhibitors, and YAP knockdown induced expression of galectin‐3 and BIM. Conclusions and Implications Stimulation of cardiac β‐adrenoceptors activated the Mst1/Hippo pathway leading to YAP hyper‐phosphorylation with enhanced expression of galectin‐3 and BIM. This signalling pathway would have therapeutic potential. Linked Articles This article is part of a themed section on Adrenoceptors—New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc
Collapse
Affiliation(s)
- Wei-Bo Zhao
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Qun Lu
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Cardiovascular Medicine, First Hospital and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - My-Nhan Nguyen
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Yidan Su
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University, Geelong, Australia
| | - Li-Na Wang
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Hou-Yuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Cardiovascular Medicine, First Hospital and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
177
|
Du XJ, Kiriazis H, Nguyen MN, Ziemann M, Zhao WB. Reply to "Letter to the Editor: Not all modified citrus pectins are the same: size does matter". Am J Physiol Heart Circ Physiol 2019; 316:H1234-H1235. [PMID: 31070460 DOI: 10.1152/ajpheart.00197.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute , Melbourne , Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute , Melbourne , Australia
| | - My-Nhan Nguyen
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute , Melbourne , Australia.,Division of Cardiovascular Medicine, John Radcliffe Hospital, University of Oxford , Oxford , United Kingdom
| | - Mark Ziemann
- School of Life and Environmental Sciences, Deakin University , Melbourne , Australia
| | - Wei-Bo Zhao
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute , Melbourne , Australia
| |
Collapse
|
178
|
Sun Z, Wang Z, Li L, Yan J, Shao C, Bao Z, Jing L, Pang Q, Geng Y, Zhang L. RAGE/galectin-3 yields intraplaque calcification transformation via sortilin. Acta Diabetol 2019; 56:457-472. [PMID: 30603868 DOI: 10.1007/s00592-018-1273-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/10/2018] [Indexed: 12/23/2022]
Abstract
AIMS Macrocalcification and microcalcification present different clinical risks, but the regulatory of their formation was unclear. Therefore, this study explored the underlying mechanisms of macrocalcification and microcalcification in diabetes mellitus. METHODS Anterior tibial arteries of amputated diabetic feet were collected. According to the calcium content, patients were divided into less-calcification group and more-calcification group. And calcification morphology in plaques was observed. For further study, an in vivo mouse diabetic atherosclerosis model and an in vitro primary mouse aortic smooth muscle cell model were established. After the receptors for AGEs (RAGE) or galectin-3 were silenced, calcified nodule sizes and sortilin expression were determined. Scanning electron microscopy (SEM) was performed to detect the aggregation of matrix vesicles with the inhibition or promotion of sortilin. RESULTS Both macro- and microcalcification were found in human anterior tibial artery plaques. Macrocalcification formed after the silencing of RAGE, and microcalcification formed after the silencing of galectin-3. In the process of RAGE- or galcetin-3-induced calcification, sortilin played an important role downstream. SEM showed that sortilin promoted the aggregation of MVs in the early stage of calcification and formed larger calcified nodules. CONCLUSION RAGE downregulated sortilin and then transmitted microcalcification signals, whereas galectin-3 upregulated sortilin, which accelerated the aggregation of MVs in the early stage of calcification and mediated the formation of macrocalcifications, These data illustrate the progression of two calcification types and suggest sortilin as a potential target for early intervention of calcification and as an effective biomarker for the assessment of long-term clinical risk and prognosis.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/metabolism
- Amputation, Surgical
- Animals
- Aorta/metabolism
- Aorta/pathology
- Blood Proteins
- Cells, Cultured
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/surgery
- Diabetic Foot/pathology
- Diabetic Foot/surgery
- Galectin 3/physiology
- Galectins
- Gene Expression Regulation/drug effects
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- RNA Interference
- RNA, Small Interfering/pharmacology
- Receptor for Advanced Glycation End Products/physiology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Streptozocin
- Tibial Arteries/metabolism
- Tibial Arteries/pathology
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China.
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Zhengyang Bao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Lele Jing
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Qiwen Pang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Yue Geng
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212001, Zhenjiang, China
| |
Collapse
|
179
|
Ghorbani A, Ho JE. Reply. J Am Coll Cardiol 2019; 73:1875-1876. [DOI: 10.1016/j.jacc.2019.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 02/11/2019] [Indexed: 11/29/2022]
|
180
|
Kirk JA, de Boer RA. Moving galectin-3 closer to the goal line. Am J Physiol Heart Circ Physiol 2019; 316:H580-H582. [PMID: 30575435 PMCID: PMC6415817 DOI: 10.1152/ajpheart.00761.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 11/22/2022]
Affiliation(s)
- Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine , Maywood, Illinois
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen , Groningen , The Netherlands
| |
Collapse
|
181
|
Berezin AE. Prognostication of clinical outcomes in diabetes mellitus: Emerging role of cardiac biomarkers. Diabetes Metab Syndr 2019; 13:995-1003. [PMID: 31336558 DOI: 10.1016/j.dsx.2019.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) remains substantial health problem and one of the most prevalent metabolic diseases worldwide. The impact of T2DM on CV mortality and morbidity is embedded through a nature evolution of the disease and is modulated by numerous risk factors, such as hypertension, obesity, dyslipidemia. There is large body of evidence regarding use of the cardiac biomarkers to risk stratification at higher CV risk individuals who belongs to general population and cohort with established CV disease. Although T2DM patients have higher incidence of cardiac and vascular complications than the general population, whether cardiac biomarkers would be effective to risk stratification of the T2DM is not fully understood. The aim of the review is to summarize our knowledge regarding clinical implementation of cardiac biomarkers in risk assessment for T2DM patients. The role of natriuretic peptides, soluble ST2, galectin-3, growth differentiation factor-15, and cardiac troponins are widely discussed.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, Medical University of Zaporozhye, Mayakovsky av., 25, Zaporozhye, 69035, Ukraine.
| |
Collapse
|
182
|
Stegmayr J, Zetterberg F, Carlsson MC, Huang X, Sharma G, Kahl-Knutson B, Schambye H, Nilsson UJ, Oredsson S, Leffler H. Extracellular and intracellular small-molecule galectin-3 inhibitors. Sci Rep 2019; 9:2186. [PMID: 30778105 PMCID: PMC6379368 DOI: 10.1038/s41598-019-38497-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/27/2018] [Indexed: 01/04/2023] Open
Abstract
Galectin-3 is a carbohydrate binding protein which has important roles in cancer and immunity. Potent galectin-3 inhibitors have been synthesized, for experimental purposes and potential clinical use. As galectin-3 is implicated in both intra- and extracellular activities, permeability of galectin-3 inhibitors is an important parameter determining biological effects. We compared the cellular uptake of galectin-3 inhibitors and their potency in the intracellular or extracellular space. The inhibitors differed in their polar surface area (PSA), but had similar affinities for galectin-3. Using a well-established permeability assay, we confirmed that the uptake was significantly higher for the inhibitor with the lowest PSA, as expected. To analyze intracellular activity of the inhibitors, we developed a novel assay based on galectin-3 accumulation around damaged intracellular vesicles. The results show striking differences between the inhibitors intracellular potency, correlating with their PSAs. To test extracellular activity of the inhibitors, we analyzed their potency to block binding of galectin-3 to cell surfaces. All inhibitors were equally able to block galectin-3 binding to cells and this was proportional to their affinity for galectin-3. These inhibitors may serve as useful tools in exploring biological roles of galectin-3 and may further our understanding of intracellular versus extracellular roles of galectin-3.
Collapse
Affiliation(s)
- John Stegmayr
- Department of Laboratory Medicine, Lund University, 22100, Lund, Sweden.
| | | | - Michael C Carlsson
- Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen N, Denmark.,Agilent Technologies Denmark ApS, 2600, Glostrup, Denmark
| | - Xiaoli Huang
- Department of Biology, Lund University, 22100, Lund, Sweden.,Xintela AB, 22381, Lund, Sweden
| | - Gunjan Sharma
- Department of Laboratory Medicine, Lund University, 22100, Lund, Sweden
| | | | | | - Ulf J Nilsson
- Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Stina Oredsson
- Department of Biology, Lund University, 22100, Lund, Sweden
| | - Hakon Leffler
- Department of Laboratory Medicine, Lund University, 22100, Lund, Sweden.
| |
Collapse
|
183
|
Singh M, Hanis CL, Redline S, Ballantyne CM, Hamzeh I, Aguilar D. Sleep apnea and galectin-3: possible sex-specific relationship. Sleep Breath 2019; 23:1107-1114. [PMID: 30721387 DOI: 10.1007/s11325-019-01788-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/10/2019] [Accepted: 01/23/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Sleep apnea is associated with increased risk of cardiovascular disease. Elevated plasma galectin-3 levels, a biomarker associated with myocardial fibrosis, are also associated with adverse cardiovascular events, including heart failure. Our objective was to determine the relationship between severity of sleep apnea and plasma levels of galectin-3 and to determine whether this relationship was modified by sex. METHODS We performed a cross-sectional study of 471 Mexican Americans from Starr County, TX who underwent an overnight, in-home sleep evaluation, and plasma measurement of galectin-3. Severity of sleep apnea was based on apnea hypopnea index (AHI). Multivariable linear regression modeling was used to determine the association between categories of sleep apnea and galectin-3. We also tested for interactions by sex. RESULTS The mean age was 53 years, and 74% of the cohort was female. The prevalence of moderate to severe sleep apnea (AHI > 15 apnea-hypopnea events per hour) was 36.7%. Moderate to severe sleep apnea was associated with increased levels of galectin-3 in the entire population, but we identified a statistically significant interaction between galectin-3 levels and category of sleep apnea by sex (p for interaction = 0.02). Plasma galectin levels were significantly higher in women with moderate or severe sleep apnea than women with no/mild sleep apnea (multivariable adjusted p < 0.001), but not in men (p = 0.5). CONCLUSIONS Sleep apnea is associated elevated galectin-3 levels in women but not men. Our findings highlight a possible sex-specific relationship between sleep apnea and galectin-3, a biomarker of potential myocardial fibrosis that has been associated with increased cardiovascular risk.
Collapse
Affiliation(s)
- Mohita Singh
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Craig L Hanis
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, 1200 Pressler Street, Suite E431, Houston, TX, 77030, USA
| | - Susan Redline
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christie M Ballantyne
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ihab Hamzeh
- Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - David Aguilar
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, 1200 Pressler Street, Suite E431, Houston, TX, 77030, USA.
| |
Collapse
|
184
|
de Boer RA, De Keulenaer G, Bauersachs J, Brutsaert D, Cleland JG, Diez J, Du XJ, Ford P, Heinzel FR, Lipson KE, McDonagh T, Lopez-Andres N, Lunde IG, Lyon AR, Pollesello P, Prasad SK, Tocchetti CG, Mayr M, Sluijter JPG, Thum T, Tschöpe C, Zannad F, Zimmermann WH, Ruschitzka F, Filippatos G, Lindsey ML, Maack C, Heymans S. Towards better definition, quantification and treatment of fibrosis in heart failure. A scientific roadmap by the Committee of Translational Research of the Heart Failure Association (HFA) of the European Society of Cardiology. Eur J Heart Fail 2019; 21:272-285. [PMID: 30714667 PMCID: PMC6607480 DOI: 10.1002/ejhf.1406] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a pivotal player in heart failure development and progression. Measurements of (markers of) fibrosis in tissue and blood may help to diagnose and risk stratify patients with heart failure, and its treatment may be effective in preventing heart failure and its progression. A lack of pathophysiological insights and uniform definitions has hampered the research in fibrosis and heart failure. The Translational Research Committee of the Heart Failure Association discussed several aspects of fibrosis in their workshop. Early insidious perturbations such as subclinical hypertension or inflammation may trigger first fibrotic events, while more dramatic triggers such as myocardial infarction and myocarditis give rise to full blown scar formation and ongoing fibrosis in diseased hearts. Aging itself is also associated with a cardiac phenotype that includes fibrosis. Fibrosis is an extremely heterogeneous phenomenon, as several stages of the fibrotic process exist, each with different fibrosis subtypes and a different composition of various cells and proteins — resulting in a very complex pathophysiology. As a result, detection of fibrosis, e.g. using current cardiac imaging modalities or plasma biomarkers, will detect only specific subforms of fibrosis, but cannot capture all aspects of the complex fibrotic process. Furthermore, several anti‐fibrotic therapies are under investigation, but such therapies generally target aspecific aspects of the fibrotic process and suffer from a lack of precision. This review discusses the mechanisms and the caveats and proposes a roadmap for future research.
Collapse
Affiliation(s)
- Rudolf A de Boer
- University Medical Center Groningen, University of Groningen, Department of Cardiology, Groningen, The Netherlands
| | | | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Dirk Brutsaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - John G Cleland
- Robertson Centre for Biostatistics & Clinical Trials, University of Glasgow, Glasgow, UK
| | - Javier Diez
- Program of Cardiovascular Diseases, Center for Applied Medical Research, Departments of Nephrology, and Cardiology and Cardiac Surgery, University Clinic, University of Navarra, Pamplona, Spain
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | | | - Frank R Heinzel
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | | | | | - Natalia Lopez-Andres
- Cardiovascular Translational Research, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Publica de Navarra, Idisna, Spain
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Alexander R Lyon
- Royal Brompton Hospital, and Imperial College London, London, UK
| | | | | | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Manuel Mayr
- The James Black Centre, King's College, University of London, London, UK
| | - Joost P G Sluijter
- University Medical Centre Utrecht, Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Utrecht, Utrecht, The Netherlands
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany.,DZHK (German Center for Cardiovascular Research) partner site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Campus Virchow-Klinikum, Charite Universitaetsmedizin Berlin, Berlin, Germany
| | - Faiez Zannad
- Centre d'Investigation Clinique, CHU de Nancy, Nancy, France
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research) partner site Göttingen, Göttingen, Germany
| | - Frank Ruschitzka
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Gerasimos Filippatos
- Heart Failure Unit, Department of Cardiology, School of Medicine, Athens University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| | - Merry L Lindsey
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center and Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | - Christoph Maack
- Comprehensive Heart Failure Centre, University and University Hospital Würzburg, Würzburg, Germany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium.,The Netherlands Heart Institute, Nl-HI, Utrecht, The Netherlands
| |
Collapse
|
185
|
Zhong X, Qian X, Chen G, Song X. The role of galectin-3 in heart failure and cardiovascular disease. Clin Exp Pharmacol Physiol 2019; 46:197-203. [PMID: 30372548 DOI: 10.1111/1440-1681.13048] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Xiao Zhong
- Cardiovascular Center; The Fourth Affiliated Hospital; Harbin Medical University; Harbin China
| | - Xiaoqian Qian
- Department of Nephrology; Xin Hua Hospital Affiliated; Shanghai Jiao Tong University School of Medicine; Shanghai China
| | - Guangping Chen
- Department of Physiology; Emory University School of Medicine; Atlanta Georgia
| | - Xiang Song
- Cardiovascular Center; The Fourth Affiliated Hospital; Harbin Medical University; Harbin China
| |
Collapse
|
186
|
Ghorbani A, Bhambhani V, Christenson RH, Meijers WC, de Boer RA, Levy D, Larson MG, Ho JE. Longitudinal Change in Galectin-3 and Incident Cardiovascular Outcomes. J Am Coll Cardiol 2018; 72:3246-3254. [PMID: 30573026 PMCID: PMC6516745 DOI: 10.1016/j.jacc.2018.09.076] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/12/2018] [Accepted: 09/22/2018] [Indexed: 11/18/2022]
Abstract
BACKGROUND Galectin-3 (Gal-3) has been associated with heart failure (HF) and poor cardiovascular outcomes. However, the effect of longitudinal changes in Gal-3 on clinical outcomes remains unclear. OBJECTIVES The authors sought to study clinical determinants of change in Gal-3 among community-dwelling individuals. Further, they sought to examine the role of serial Gal-3 measurements in predicting risk of future HF, cardiovascular disease (CVD), and mortality. METHODS A total of 2,477 participants in the Framingham Heart Study Offspring cohort underwent measurement of plasma Gal-3 levels at 2 examinations (1995 to 1998 and 2005 to 2008). Linear regression models were used to examine clinical correlates of change in Gal-3. Proportional hazards models were used to relate future clinical outcomes with change in Gal-3. RESULTS The following clinical correlates were associated with greater longitudinal increases in Gal-3 levels: age, female sex, hypertension, diabetes, body mass index, interim development of chronic kidney disease, and HF (p < 0.0001 for all in multivariable model). Change in Gal-3 was associated with future HF (hazard ratio [HR]: 1.39 per 1-SD increase; 95% confidence interval [CI]: 1.13 to 1.71), CVD (HR: 1.29; 95% CI: 1.11 to 1.51), and all-cause mortality (HR: 1.30; 95% CI: 1.17 to 1.46). Change in Gal-3 was associated with both HF with preserved as well as reduced ejection fraction (p < 0.05 for both). CONCLUSIONS Longitudinal changes in Gal-3 are associated with traditional cardiovascular risk factors and renal disease. In turn, change in Gal-3 predicts future HF, CVD, and mortality in the community. Future studies are needed to determine whether serial Gal-3 measures may be useful in disease prevention.
Collapse
Affiliation(s)
- Anahita Ghorbani
- Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, Connecticut
| | - Vijeta Bhambhani
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts; Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Robert H Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wouter C Meijers
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daniel Levy
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, Massachusetts; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Martin G Larson
- Framingham Heart Study, Framingham, Massachusetts; Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Jennifer E Ho
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts; Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
187
|
Galectin 3 inhibition attenuates renal injury progression in cisplatin-induced nephrotoxicity. Biosci Rep 2018; 38:BSR20181803. [PMID: 30455396 PMCID: PMC6435560 DOI: 10.1042/bsr20181803] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/12/2018] [Accepted: 11/15/2018] [Indexed: 12/22/2022] Open
Abstract
Nephrotoxicity is a major toxic effect in chemotherapy, which constitutes up to 60% of hospitalized acute kidney injury (AKI). Very few treatment options exist to slow the transition from AKI to subsequent chronic kidney diseases (CKD). Here, we demonstrate that galectin-3 (Gal-3), a β-galactoside binding lectin that plays an important role in kidney fibrosis and renal failure, is one of the key factors for renal injury progression. Ectopic overexpression of Gal-3 significantly decreased the viability of HEK293, simultaneously inducing of cell cycle arrest and apoptosis. However, inhibition of Gal-3, mediated by modified citrus pectin (MCP), predominantly antagonized the pro-apoptotic effects. Mice were pre-treated with normal or 1% MCP-supplemented drinking water 1 week before cisplatin injection. Analyses of serum creatinine and renal tissue damage indicated that MCP-treated mice demonstrated increased renal function and attenuated renal fibrosis after cisplatin-induced injury. MCP-treated mice also demonstrated decreased renal fibrosis and apoptosis, as revealed by masson trichrome staining and Western blot analysis of cleaved caspase-3. Additionally, the protective role of Gal-3 inhibition in the kidney injury was shown to be mediated by protein kinase C α (PKC-α), which promoted cell apoptosis and collagen I synthesis in HEK293 cells. These results demonstrated the potential Gal-3 and PKC-α as therapeutic targets for the treatment of AKI and CKD.
Collapse
|
188
|
Zeng N, Wang A, Zhong C, Zheng X, Zhu Z, Xu T, Peng Y, Peng H, Li Q, Ju Z, Geng D, Zhang Y, He J. Association of serum galectin-3 with risks of death and vascular events in acute ischaemic stroke patients: the role of hyperglycemia. Eur J Neurol 2018; 26:415-421. [PMID: 30414289 DOI: 10.1111/ene.13856] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/05/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND PURPOSE Whether the association between galectin-3 and stroke outcome is modified by fasting plasma glucose (FPG) is unknown. The aim was to evaluate the prognostic effect of galectin-3 amongst ischaemic stroke patients stratified by FPG. METHODS In all, 3082 ischaemic stroke patients were included in this study and serum galectin-3 was tested at baseline. The primary outcome was a composite outcome of death and vascular events, and secondary outcomes were death, stroke recurrence and vascular events within 1 year after stroke. RESULTS Increased galectin-3 was significantly associated with the primary outcome, stroke recurrence and vascular events in the patients with hyperglycemia but not in those with normoglycemia (P for interaction < 0.05 for all). The multivariate-adjusted hazard ratios (95% confidence intervals) were 1.72 (1.05-2.84), 2.64 (1.14-6.12) and 2.68 (1.33-5.38) for the primary outcome, stroke recurrence and vascular events, respectively. A linear association between galectin-3 and the primary outcome was observed in hyperglycemic patients (P for linearity = 0.007). CONCLUSION Increased galectin-3 was associated with the primary outcome, stroke recurrence and vascular events within 1 year after stroke in the patients with hyperglycemia, suggesting that galectin-3 may be an important prognostic factor for ischaemic stroke patients with hyperglycemia.
Collapse
Affiliation(s)
- N Zeng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - A Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - C Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - X Zheng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Z Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - T Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Y Peng
- Department of Neurology, Affiliated Hospital of North China University of Science and Technology, Hebei, China
| | - H Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Q Li
- Department of Epidemiology, School of Public Health, Taishan Medical College, Shandong, China
| | - Z Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Tongliao, Inner Mongolia, China
| | - D Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical College, Jiangsu, China
| | - Y Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - J He
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| |
Collapse
|
189
|
Zheng Y, Feng W, Wang YJ, Sun Y, Shi G, Yu Q. Galectins as potential emerging key targets in different types of leukemia. Eur J Pharmacol 2018; 844:73-78. [PMID: 30452910 DOI: 10.1016/j.ejphar.2018.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/20/2018] [Accepted: 11/14/2018] [Indexed: 01/11/2023]
Abstract
Galectins are carbohydrate-binding proteins and these have very high affinity for β-galactoside containing glycoproteins and glycolipids. Amongst sixteen types of galectin, the role of galectin 1, 3, 9 and 12 is defined in the development and progression of different types of leukemia including acute myeloid leukemia, acute promyelocytic leukemia, B-cell precursor acute lymphoblastic leukemia, adult T cell leukemia and chronic lymphocytic leukemia. There are multiple mechanisms through which these galectins may affect tumor proliferation. These may include increased production of tumor resistance conferring proteins such as multidrug resistance (MDR-1) and myeloid cell leukemia (MCL-1). Moreover, galectin-9 may act on Tim-3 receptors present on the circulating CD8+ T cells to impair immune system function and the latter provide an ideal environment for the proliferation of leukemic cells. The present review describes the role and mechanisms involved in galectin-mediated development and progression of different types of leukemia.
Collapse
Affiliation(s)
- Yan Zheng
- The Department of Anesthesia, China-Japan Union Hospital of Jilin University, China.
| | - Wei Feng
- The Department of Anesthesia, China-Japan Union Hospital of Jilin University, China
| | - Yu-Juan Wang
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Yan Sun
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Guang Shi
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| | - Qiong Yu
- The Department of Hematology and Oncology, The Second Hospital of Jilin University, China.
| |
Collapse
|
190
|
Laaf D, Bojarová P, Elling L, Křen V. Galectin-Carbohydrate Interactions in Biomedicine and Biotechnology. Trends Biotechnol 2018; 37:402-415. [PMID: 30413271 DOI: 10.1016/j.tibtech.2018.10.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/25/2018] [Accepted: 10/02/2018] [Indexed: 12/23/2022]
Abstract
Cellular communication events are mediated by interactions between cell-surface sugars and lectins, which are carbohydrate-binding proteins. Galectins are β-galactosyl-binding lectins that bridge molecules by their sugar moieties, forming a signaling and adhesion network. Severe changes in glycosylation and galectin expression accompany major processes in oncogenesis, cardiovascular disorders, and other pathologies, making galectins attractive therapeutic targets. Here we discuss advanced strategies of chemo-enzymatic carbohydrate synthesis for creating lead glycomimetics and (neo-)glycoconjugates for galectin-1 and -3 targeting in biomedicine and biotechnology. We will describe the challenges and bottlenecks on the route into biomedical and biotechnological practice and present the first clinical candidates. The coming era will see an exciting translation of selective well-defined high-affinity galectin ligands from bench to bedside.
Collapse
Affiliation(s)
- Dominic Laaf
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, D-52074 Aachen, Germany; Equally contributing authors
| | - Pavla Bojarová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4, Czech Republic; Equally contributing authors
| | - Lothar Elling
- Laboratory for Biomaterials, Institute for Biotechnology and Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Pauwelsstraße 20, D-52074 Aachen, Germany.
| | - Vladimír Křen
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Prague 4, Czech Republic.
| |
Collapse
|
191
|
Melendez GC, Chen Q, Lesnefsky EJ. Metformin as a modulator of myocardial fibrosis postmyocardial infarction via regulation of cardiomyocyte-fibroblast crosstalk. Transl Res 2018; 199:1-3. [PMID: 30244699 PMCID: PMC9113666 DOI: 10.1016/j.trsl.2018.05.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 12/01/2022]
Affiliation(s)
- Giselle C Melendez
- Department of Internal Medicine, Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Pathology, Comparative Medicine; Wake Forest School of Medicine, Winston-Salem, North Carolina.
| | - Qun Chen
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Internal Medicine, Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Biochemistry, and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia; Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia; The Medical Service of the McGuire Veterans Affairs Medical Center in Richmond, Virginia.
| |
Collapse
|
192
|
Du W, Piek A, Schouten EM, de Kolk CWV, Mueller C, Mebazaa A, A.Voors A, de Boer RA, Silljé HH. Plasma levels of heart failure biomarkers are primarily a reflection of extracardiac production. Theranostics 2018; 8:4155-4169. [PMID: 30128044 PMCID: PMC6096401 DOI: 10.7150/thno.26055] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022] Open
Abstract
Plasma heart failure (HF) biomarkers, like natriuretic peptides, are important in diagnosis, prognosis and HF treatment. Several novel HF biomarkers have been identified, including Gal-3, GDF-15 and TIMP-1, but their clinical potential remains vague. Here we investigated plasma biomarker levels in relation to tissue expression and structural and functional cardiac changes. Methods: Cardiac remodeling, cardiac function, and plasma and tissue biomarker levels were investigated in mice after myocardial infarction induced by temporal and permanent LAD ligation (tLAD and pLAD). In addition, a pressure overload model induced by transverse aortic constriction (TAC) and an obese/hypertensive HFpEF-like mouse model were investigated. Results: Plasma levels of ANP and its cardiac expression were strictly associated with cardiac remodeling and function. Gal-3, GDF-15 and TIMP-1 cardiac expressions were also related to cardiac remodeling and function, but not their plasma levels. Only directly after myocardial infarction could elevated plasma levels of Gal-3 and TIMP-1 be detected. Eight weeks after infarction, plasma levels were not elevated despite enhanced cardiac expression and low EF (18.3±3.3%, pLAD). Plasma levels of TIMP-1 and GDF-15 were elevated after TAC, but this also correlated with increased lung expression and congestion. In obese-hypertensive mice, elevated plasma levels of Gal-3, GDF-15 and TIMP1 were associated with increased adipose tissue expression and not with cardiac function. Conclusions: The Gal-3, GDF-15 and TIMP-1 plasma pool levels are hardly influenced by dynamic changes in cardiac expression. These biomarkers are not specific for indices of cardiac remodeling, but predominantly reflect stress in other affected tissues and hence provide health information beyond the heart.
Collapse
|
193
|
Lam CSP, Voors AA, de Boer RA, Solomon SD, van Veldhuisen DJ. Heart failure with preserved ejection fraction: from mechanisms to therapies. Eur Heart J 2018; 39:2780-2792. [DOI: 10.1093/eurheartj/ehy301] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022] Open
Affiliation(s)
- Carolyn S P Lam
- National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, Hanzeplein 1, Groningen, the Netherlands
- Duke-National University of Singapore Medical School, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, Hanzeplein 1, Groningen, the Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, Hanzeplein 1, Groningen, the Netherlands
| | - Scott D Solomon
- Harvard Medical School, Harvard University, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, 75 Francis St, Boston, MA, USA
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, AB31, Hanzeplein 1, Groningen, the Netherlands
| |
Collapse
|
194
|
Galectin-3 in Atrial Fibrillation: Mechanisms and Therapeutic Implications. Int J Mol Sci 2018; 19:ijms19040976. [PMID: 29587379 PMCID: PMC5979515 DOI: 10.3390/ijms19040976] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/15/2018] [Accepted: 03/23/2018] [Indexed: 12/30/2022] Open
Abstract
Maintenance of atrial fibrillation is a complex mechanism, including extensive electrical and structural remodeling of the atria which involves progressive fibrogenesis. Galectin-3 is a biomarker of fibrosis, and, thus, may be involved in atrial remodeling in atrial fibrillation patients. We review the role of galectin-3 in AF mechanisms and its potential therapeutic implications.
Collapse
|