1
|
Contaldi C, D’Aniello C, Panico D, Zito A, Calabrò P, Di Lorenzo E, Golino P, Montesarchio V. Cancer-Therapy-Related Cardiac Dysfunction: Latest Advances in Prevention and Treatment. Life (Basel) 2025; 15:471. [PMID: 40141815 PMCID: PMC11944213 DOI: 10.3390/life15030471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
The increasing efficacy of cancer therapies has significantly improved survival rates, but it has also highlighted the prevalence of cancer-therapy-related cardiac dysfunction (CTRCD). This review provides a comprehensive overview of the identification, monitoring, and management of CTRCD, a condition resulting from several treatments, such as anthracyclines, HER2-targeted therapies, target therapies, and radiotherapy. The paper includes a discussion of the mechanisms of CTRCD associated with various cancer treatments. Early detection through serum biomarkers and advanced imaging techniques is crucial for effective monitoring and risk stratification. Preventive strategies include pharmacological interventions such as ACE inhibitors/angiotensin receptor blockers, beta-blockers, and statins. Additionally, novel agents like sacubitril/valsartan, sodium-glucose co-transporter type 2 inhibitors, and vericiguat show promise in managing left ventricular dysfunction. Lifestyle modifications, including structured exercise programs and optimized nutritional strategies, further contribute to cardioprotection. The latest treatments for both asymptomatic and symptomatic CTRCD across its various stages are also discussed. Emerging technologies, including genomics, artificial intelligence, novel biomarkers, and gene therapy, are paving the way for personalized approaches to CTRCD prevention and treatment. These advancements hold great promise for improving long-term outcomes in cancer patients by minimizing cardiovascular complications.
Collapse
Affiliation(s)
- Carla Contaldi
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Carmine D’Aniello
- Division of Medical Oncology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Domenico Panico
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Andrea Zito
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | - Emilio Di Lorenzo
- Department of Cardiology, AORN dei Colli-Monaldi Hospital, 80131 Naples, Italy
| | - Paolo Golino
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy
| | | |
Collapse
|
2
|
Junyent M, Noori H, De Schepper R, Frajdenberg S, Elsaigh RKAH, McDonald PH, Duckett D, Maudsley S. Unravelling Convergent Signaling Mechanisms Underlying the Aging-Disease Nexus Using Computational Language Analysis. Curr Issues Mol Biol 2025; 47:189. [PMID: 40136443 PMCID: PMC11941692 DOI: 10.3390/cimb47030189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/12/2025] [Accepted: 03/08/2025] [Indexed: 03/27/2025] Open
Abstract
Multiple lines of evidence suggest that multiple pathological conditions and diseases that account for the majority of human mortality are driven by the molecular aging process. At the cellular level, aging can largely be conceptualized to comprise the progressive accumulation of molecular damage, leading to resultant cellular dysfunction. As many diseases, e.g., cancer, coronary heart disease, Chronic obstructive pulmonary disease, Type II diabetes mellitus, or chronic kidney disease, potentially share a common molecular etiology, then the identification of such mechanisms may represent an ideal locus to develop targeted prophylactic agents that can mitigate this disease-driving mechanism. Here, using the input of artificial intelligence systems to generate unbiased disease and aging mechanism profiles, we have aimed to identify key signaling mechanisms that may represent new disease-preventing signaling pathways that are ideal for the creation of disease-preventing chemical interventions. Using a combinatorial informatics approach, we have identified a potential critical mechanism involving the recently identified kinase, Dual specificity tyrosine-phosphorylation-regulated kinase 3 (DYRK3) and the epidermal growth factor receptor (EGFR) that may function as a regulator of the pathological transition of health into disease via the control of cellular fate in response to stressful insults.
Collapse
Affiliation(s)
- Marina Junyent
- Receptor Biology Lab., University of Antwerp, 2610 Wilrijk, Belgium; (M.J.); (H.N.); (R.D.S.); (S.F.); (R.K.A.H.E.)
- IMIM, Hospital del Mar Research Institute, 08003 Barcelona, Spain
| | - Haki Noori
- Receptor Biology Lab., University of Antwerp, 2610 Wilrijk, Belgium; (M.J.); (H.N.); (R.D.S.); (S.F.); (R.K.A.H.E.)
- Department of Chemistry, KU Leuven, Oude Markt 13, 3000 Leuven, Belgium
| | - Robin De Schepper
- Receptor Biology Lab., University of Antwerp, 2610 Wilrijk, Belgium; (M.J.); (H.N.); (R.D.S.); (S.F.); (R.K.A.H.E.)
| | - Shanna Frajdenberg
- Receptor Biology Lab., University of Antwerp, 2610 Wilrijk, Belgium; (M.J.); (H.N.); (R.D.S.); (S.F.); (R.K.A.H.E.)
| | | | - Patricia H. McDonald
- Lexicon Pharmaceuticals Inc., 2445 Technology Forest Blvd Fl 1, The Woodlands, TX 77381, USA;
| | - Derek Duckett
- Department of Drug Discovery, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| | - Stuart Maudsley
- Receptor Biology Lab., University of Antwerp, 2610 Wilrijk, Belgium; (M.J.); (H.N.); (R.D.S.); (S.F.); (R.K.A.H.E.)
- Department of Drug Discovery, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA;
| |
Collapse
|
3
|
Castro C, Delwarde C, Shi Y, Roh J. Geroscience in heart failure: the search for therapeutic targets in the shared pathobiology of human aging and heart failure. THE JOURNAL OF CARDIOVASCULAR AGING 2025; 5:10.20517/jca.2024.15. [PMID: 40297496 PMCID: PMC12036312 DOI: 10.20517/jca.2024.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Age is a major risk factor for heart failure, but one that has been historically viewed as non-modifiable. Emerging evidence suggests that the biology of aging is malleable, and can potentially be intervened upon to treat age-associated chronic diseases, such as heart failure. While aging biology represents a new frontier for therapeutic target discovery in heart failure, the challenges of translating Geroscience research to the clinic are multifold. In this review, we propose a strategy that prioritizes initial target discovery in human biology. We review the rationale for starting with human omics, which has generated important insights into the shared (patho)biology of human aging and heart failure. We then discuss how this knowledge can be leveraged to identify the mechanisms of aging biology most relevant to heart failure. Lastly, we provide examples of how this human-first Geroscience approach, when paired with rigorous functional assessments in preclinical models, is leading to early-stage clinical development of gerotherapeutic approaches for heart failure.
Collapse
Affiliation(s)
- Claire Castro
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Constance Delwarde
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Yanxi Shi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
4
|
Cools JMT, Goovaerts BK, Feyen E, Van den Bogaert S, Fu Y, Civati C, Van Fraeyenhove J, Tubeeckx MRL, Ott J, Nguyen L, Wülfers EM, Van Berlo B, De Vries AAF, Vandersickel N, Pijnappels DA, Audenaert D, Roderick HL, De Winter H, De Keulenaer GW, Segers VFM. Small-molecule-induced ERBB4 activation to treat heart failure. Nat Commun 2025; 16:576. [PMID: 39794341 PMCID: PMC11724075 DOI: 10.1038/s41467-024-54908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025] Open
Abstract
Heart failure is a common and deadly disease requiring new treatments. The neuregulin-1/ERBB4 pathway offers cardioprotective benefits, but using recombinant neuregulin-1 as therapy has limitations due to the need for intravenous delivery and lack of receptor specificity. We hypothesize that small-molecule activation of ERBB4 could protect against heart damage and fibrosis. To test this, we conduct a screening of 10,240 compounds and identify eight structurally similar ones (EF-1 to EF-8) that induce ERBB4 dimerization, with EF-1 being the most effective. EF-1 reduces cell death and hypertrophy in cardiomyocytes and decreases collagen production in cardiac fibroblasts in an ERBB4-dependent manner. In wild-type mice, EF-1 inhibits angiotensin-II-induced fibrosis in males and females and reduces heart damage caused by doxorubicin and myocardial infarction in females, but not in Erbb4-null mice. This study shows that small-molecule ERBB4 activation is feasible and may lead to a novel class of drugs for treating heart failure.
Collapse
Grants
- This work was supported by a Geconcerteerde onderzoeksactie grant (GOA, PID36444) of the University of Antwerp; by a Senior Clinical Investigator fellowship (to V.F.S.), a PhD fellowship (to J.MT.C. and C.C.), and research grants of the Fund for Scientific Research Flanders (Application numbers 1842219N, G021019N, G021420N, 1S49323N, and 11PBU24N); VLIR/iBOF Grant 20-VLIR-iBOF-027 (to N.V., V.F.S., H.L.R., and G.W.D.K.).
Collapse
Affiliation(s)
- Julie M T Cools
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Bo K Goovaerts
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Eline Feyen
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | - Yile Fu
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Céline Civati
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | | | - Jasper Ott
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | - Long Nguyen
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Eike M Wülfers
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg im Breisbau, Germany and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Benji Van Berlo
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Antoine A F De Vries
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dominique Audenaert
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium.
| |
Collapse
|
5
|
Jie S, Wenying G, Lebo S. Dehydroevodiamine Alleviates Doxorubicin-Induced Cardiomyocyte Injury by Regulating Neuregulin-1/ErbB Signaling. Cardiovasc Ther 2024; 2024:5538740. [PMID: 39742014 PMCID: PMC11646148 DOI: 10.1155/cdr/5538740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Doxorubicin (DOX) is a widely used antitumor drug; however, its use is limited by the risk of serious cardiotoxicity. Dehydroevodiamine (DHE) is a quinazoline alkaloid which has antiarrhythmic effects. The aim of this study was to investigate the protective effect of DHE on doxorubicin-induced cardiotoxicity (DIC) and its potential mechanism. Materials and Methods: Rat H9c2 cardiomyocytes were exposed to DOX for 24 h to establish a DOX-induced cardiomyocyte injury model. DHE and ErbB inhibitor AG1478 were used to treat H9c2 cells to investigate their effects. Cell counting kit-8 (CCK-8) and lactate dehydrogenase (LDH) release assays were used to evaluate cell viability. Flow cytometry and caspase-3 activity assay were used to detect apoptosis. Western blot was used to detect the expression levels of apoptosis-related proteins and neuregulin-1 (NRG1)/ErbB pathway-related proteins. The levels of proinflammatory cytokines and markers of oxidative stress were also detected, respectively. Quantitative polymerase chain reaction (qPCR) was used to detect mRNA expression levels of hub genes. Results: DHE enhanced cardiomyocyte viability and decreased LDH release in a concentration- and time-dependent manner. DHE also significantly inhibited DOX-induced cardiomyocyte apoptosis, inflammation, and oxidative stress. Bioinformatics analysis showed that the protective mechanism of DHE against DIC was related to ErbB signaling pathway. DOX treatment significantly reduced NRG1, p-ErbB2, and p-ErbB4 protein expression levels in cardiomyocytes, while DHE pretreatment reversed this effect. ErbB inhibitor AG1478 reversed the protective effect of DHE on cardiomyocytes. Conclusion: DHE protects cardiomyocytes against DOX by regulating NRG1/ErbB pathway. DHE may be a potential agent for the prevention and treatment of DIC.
Collapse
Affiliation(s)
- Song Jie
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| | - Guo Wenying
- Department of Digestive, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| | - Sun Lebo
- Department of Cardiothoracic Surgery, Ningbo Medical Center Lihuili Hospital of Ningbo University, No. 57, Xingning Rd, Ningbo City 315041, Zhejiang Province, China
| |
Collapse
|
6
|
Banerjee S, Oguljahan B, Thompson WE, Chowdhury I. Neuregulin 1 Signaling Attenuates Tumor Necrosis Factor α-Induced Female Rat Luteal Cell Death. Endocrinology 2024; 165:bqae129. [PMID: 39312480 PMCID: PMC11456883 DOI: 10.1210/endocr/bqae129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/16/2024] [Accepted: 09/20/2024] [Indexed: 09/25/2024]
Abstract
The corpus luteum (CL) is a transient ovarian endocrine structure that maintains pregnancy in primates during the first trimester and in rodents during the entire pregnancy by producing steroid hormone progesterone (P4). CL lifespan, growth, and differentiation are tightly regulated by survival and cell death signals through luteotrophic and luteolytic factors, including the epidermal growth factor (EGF)-like factor family. Neuregulin 1 (NRG1), a member of the EGF family, mediates its effect through ErbB2/3 receptors. However, the functional role of NRG1 in luteal cells (LCs) is unknown. Thus, this study investigated the role of NRG1 and its molecular mechanism of action in rat LC. Our experimental results suggest a strong positive correlation between steroidogenic acute regulatory protein (StAR) and NRG1 expression in mid-CL and serum P4 and estrogen (E2) production. In contrast, there was a decrease in StAR and NRG1 expression and P4 and E2 production with an increase in tumor necrosis factor α (TNFα) expression in regressing CL. Further in vitro studies in LCs showed that the knockdown of endogenous Nrg1 promoted the expression of proinflammatory and proapoptotic factors and decreased prosurvival factor expression. Subsequently, treatment with exogenous TNFα under these experimental conditions profoundly elevated proinflammatory and proapoptotic factors. Further analysis demonstrated that the phosphorylation status of ErbB2/3, PI3K, Ak strain transforming or protein kinase B (Akt), and ErK1/2 was significantly inhibited under these experimental conditions, whereas the treatment of TNFα further inhibited the phosphorylation of ErbB2/3, PI3K, Akt, and ErK1/2. Collectively, these studies provide new insights into the NRG1-mediated immunomodulatory and prosurvival role in LCs, which may maintain the function of CL.
Collapse
Affiliation(s)
- Saswati Banerjee
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Babayewa Oguljahan
- Center for Laboratory Animal Resources, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Winston E Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Indrajit Chowdhury
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
7
|
Kitasato L, Yamaoka-Tojo M, Suzuki M, Nakahara S, Iwaya T, Ogiso S, Murayama Y, Hashikata T, Misawa N, Kawashima R, Oikawa J, Nakamura M, Tokui Y, Naraba J, Nishii M, Kitasato H, Ako J. Fibroblasts activation by embryonic signal switching: A novel mechanism of placental growth factor-induced cardiac remodeling. Placenta 2024; 154:129-136. [PMID: 38971073 DOI: 10.1016/j.placenta.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION Cardiac remodeling is defined as cellular interstitial changes that lead dysfunction of the heart after injury. Placental growth factor (PlGF), a member of the VEGF family, has been reported to regulate cardiac hypertrophy in hemodynamic state. We therefore analyze the function of PlGF during cardiac remodeling using cardiac cells and fibroblasts, under Angiotensin II (AngII) stimulation. METHODS PlGF overexpressed mouse embryonic fibroblasts derived from C57BL/6 mice, were made by deficient retrovirus vector, designated as C57/PlGF. Only retrovirus vector introduced C57 cells (C57/EV) were used as control. After AngII stimulation, wound scratching assay and MTT proliferation assay with or without p38 MAPK inhibitor, SB205580 were performed in retrovirally-introduced C57 cells. Reactive oxygen species (ROS) production, NF-kB activation, IL-6 and TNF-α production were also measured. Then we assessed AngII-induced cell proliferation of mouse cardiac fibroblasts (CFs) and rat primary cardiomyocytes incubating with C57/PlGF conditioned-medium. RESULTS The PlGF production in C57/PlGF were confirmed by ELISA (1093.48 ± 3.5 pg/ml, ±SE). AngII-induced cell migration, proliferation and H2O2 production were increased in C57/PlGF compared with C57/EV. SB205580 inhibited the AngII-induced cell proliferation in C57/PlGF. In C57/PlGF cells, NF-kB activation was higher, followed by up-regulation of IL-6 and TNF-α production. CFs and cardiomyocytes proliferation increased when stimulated with C57/PlGF conditioned-medium. DISCUSSION The activation of fibroblast is stimulated by PlGF signaling via p38 MAPK/NF-kB pathway accompanied by elevation of ROS and inflammatory response. Furthermore, these signals stimulate the activation of CFs and cardiomyocytes, indicating that high circulating level of PlGF have a potential to regulate cardiac remodeling.
Collapse
Affiliation(s)
- Lisa Kitasato
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan.
| | - Minako Yamaoka-Tojo
- Department of Rehabilitation, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Machika Suzuki
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Shohei Nakahara
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Toshiyuki Iwaya
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Sho Ogiso
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Yusuke Murayama
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Takehiro Hashikata
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Nonoka Misawa
- Department of Regulation Biochemistry, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Rei Kawashima
- Department of Regulation Biochemistry, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Jun Oikawa
- Department of Kitasato Clinical Research Center, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Masaki Nakamura
- Department of Laboratory Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| | - Yumi Tokui
- Department of Microbiology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku Sagamihara, Kanagawa, 252-0373, Japan; Department Environmental Microbiology, Kitasato University Graduate School of Medical Science, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Jun Naraba
- Department of Microbiology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku Sagamihara, Kanagawa, 252-0373, Japan; Department Environmental Microbiology, Kitasato University Graduate School of Medical Science, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Mototsugu Nishii
- Department of Emergency Medicine, Yokohama City University, School of Medicine, Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Hidero Kitasato
- Department of Microbiology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Minamiku Sagamihara, Kanagawa, 252-0373, Japan; Department Environmental Microbiology, Kitasato University Graduate School of Medical Science, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0373, Japan; Shibasaburo Kitasato Memorial Museum, 3199 Kitazato, Oguni, Aso, Kumamoto, 869-2505, Japan
| | - Junya Ako
- Department of Cardiovascular Medicine, Kitasato University School of Medicine, 1-15-1 Kitasato, Minamiku, Sagamihara, Kanagawa, 252-0374 Japan
| |
Collapse
|
8
|
Chen W, Gao X, Yang W, Xiao X, Pan X, Li H. Htr2b Promotes M1 Microglia Polarization and Neuroinflammation after Spinal Cord Injury via Inhibition of Neuregulin-1/ErbB Signaling. Mol Neurobiol 2024; 61:1643-1654. [PMID: 37747614 DOI: 10.1007/s12035-023-03656-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023]
Abstract
The secondary injury of spinal cord injury (SCI) is dominated by neuroinflammation, which was caused by microglia M1 polarization. This study aimed to investigate the role and mechanism of Htr2b on neuroinflammation of SCI. The BV2 and HMC3 microglia were treated with lipopolysaccharide (LPS) or interferon (IFN)-γ to simulate in vitro models of SCI. Sprague-Dawley rats were subjected to the T10 laminectomy to induce animal model of SCI. Htr2b mRNA expression was measured by qRT-PCR. The expression of Htr2b and Iba-1 was detected by western blot and immunofluorescence. The expression of inflammatory cytokines in vitro and in vivo was also measured. Kyoto Encyclopedia of Genes and Genomes (KEGG) was employed to analyze Htr2b-regulated signaling pathways. Rat behavior was analyzed by the Basso, Beattie, and Bresnahan (BBB) and inclined plane test. Rat dorsal horn tissues were stained by hematoxylin-eosin (H&E) and Nissl to measure neuron loss. Htr2b was highly expressed in LPS- and IFN-γ-treated microglia and SCI rats. SCI modeling promoted M1 microglia polarization and increased levels of inflammatory cytokines. Inhibition of Htr2b by Htr2b shRNA or RS-127445 reduced the expression of Htr2b, Iba-1, and iNOS and suppressed cytokine levels. KEGG showed that Htr2b inhibited ErbB signaling pathway. Inhibition of Htr2b increased protein expression of neuregulin-1 (Nrg-1) and p-ErbB4. Inhibition of the ErbB signaling pathway markedly reversed the effect of Htr2b shRNA on M1 microglia polarization and inflammatory cytokines. Htr2b promotes M1 microglia polarization and neuroinflammation after SCI by inhibiting Nrg-1/ErbB signaling pathway.
Collapse
Affiliation(s)
- Wenhao Chen
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China
- Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, People's Republic of China
| | - Xianlei Gao
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China
| | - Wanliang Yang
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China
- Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, People's Republic of China
| | - Xun Xiao
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China
- Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, People's Republic of China
| | - Xin Pan
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China
| | - Hao Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, No. 107, Wenhuaxi Road, Lixia District, 250012, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
9
|
Wu X, Xiao X, Fang H, He C, Wang H, Wang M, Lan P, Wang F, Du Q, Yang H. Elucidating shared biomarkers in gastroesophageal reflux disease and idiopathic pulmonary fibrosis: insights into novel therapeutic targets and the role of angelicae sinensis radix. Front Pharmacol 2024; 15:1348708. [PMID: 38414734 PMCID: PMC10897002 DOI: 10.3389/fphar.2024.1348708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Background: The etiological underpinnings of gastroesophageal reflux disease (GERD) and idiopathic pulmonary fibrosis (IPF) remain elusive, coupled with a scarcity of effective therapeutic interventions for IPF. Angelicae sinensis radix (ASR, also named Danggui) is a Chinese herb with potential anti-fibrotic properties, that holds promise as a therapeutic agent for IPF. Objective: This study seeks to elucidate the causal interplay and potential mechanisms underlying the coexistence of GERD and IPF. Furthermore, it aims to investigate the regulatory effect of ASR on this complex relationship. Methods: A two-sample Mendelian randomization (TSMR) approach was employed to delineate the causal connection between gastroesophageal reflux disease and IPF, with Phennoscanner V2 employed to mitigate confounding factors. Utilizing single nucleotide polymorphism (SNPs) and publicly available microarray data, we analyzed potential targets and mechanisms related to IPF in GERD. Network pharmacology and molecular docking were employed to explore the targets and efficacy of ASR in treating GERD-related IPF. External datasets were subsequently utilized to identify potential diagnostic biomarkers for GERD-related IPF. Results: The IVW analysis demonstrated a positive causal relationship between GERD and IPF (IVW: OR = 1.002, 95%CI: 1.001, 1.003; p < 0.001). Twenty-five shared differentially expressed genes (DEGs) were identified. GO functional analysis revealed enrichment in neural, cellular, and brain development processes, concentrated in chromosomes and plasma membranes, with protein binding and activation involvement. KEGG analysis unveiled enrichment in proteoglycan, ERBB, and neuroactive ligand-receptor interaction pathways in cancer. Protein-protein interaction (PPI) analysis identified seven hub genes. Network pharmacology analysis demonstrated that 104 components of ASR targeted five hub genes (PDE4B, DRD2, ERBB4, ESR1, GRM8), with molecular docking confirming their excellent binding efficiency. GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF (ESR1: AUCGERD = 0.762, AUCIPF = 0.725; GRM8: AUCGERD = 0.717, AUCIPF = 0.908). GRM8 and ESR1 emerged as potential diagnostic biomarkers for GERD-related IPF, validated in external datasets. Conclusion: This study establishes a causal link between GERD and IPF, identifying five key targets and two potential diagnostic biomarkers for GERD-related IPF. ASR exhibits intervention efficacy and favorable binding characteristics, positioning it as a promising candidate for treating GERD-related IPF. The potential regulatory mechanisms may involve cell responses to fibroblast growth factor stimulation and steroidal hormone-mediated signaling pathways.
Collapse
Affiliation(s)
- Xuanyu Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyu Fang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonary Diseases, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Cuifang He
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hanyue Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Miao Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peishu Lan
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quanyu Du
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Mishra N, Kant R, Kandhari K, Ammar DA, Tewari-Singh N, Pantcheva MB, Petrash JM, Agarwal C, Agarwal R. Nitrogen Mustard-Induced Ex Vivo Human Cornea Injury Model and Therapeutic Intervention by Dexamethasone. J Pharmacol Exp Ther 2024; 388:484-494. [PMID: 37474260 PMCID: PMC10801761 DOI: 10.1124/jpet.123.001760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Sulfur mustard (SM), a vesicating agent first used during World War I, remains a potent threat as a chemical weapon to cause intentional/accidental chemical emergencies. Eyes are extremely susceptible to SM toxicity. Nitrogen mustard (NM), a bifunctional alkylating agent and potent analog of SM, is used in laboratories to study mustard vesicant-induced ocular toxicity. Previously, we showed that SM-/NM-induced injuries (in vivo and ex vivo rabbit corneas) are reversed upon treatment with dexamethasone (DEX), a US Food and Drug Administration-approved, steroidal anti-inflammatory drug. Here, we optimized NM injuries in ex vivo human corneas and assessed DEX efficacy. For injury optimization, one cornea (randomly selected from paired eyes) was exposed to NM: 100 nmoles for 2 hours or 4 hours, and 200 nmoles for 2 hours, and the other cornea served as a control. Injuries were assessed 24 hours post NM-exposure. NM 100 nmoles exposure for 2 hours was found to cause optimal corneal injury (epithelial thinning [∼69%]; epithelial-stromal separation [6-fold increase]). In protein arrays studies, 24 proteins displayed ≥40% change in their expression in NM exposed corneas compared with controls. DEX administration initiated 2 hours post NM exposure and every 8 hours thereafter until 24 hours post-exposure reversed NM-induced corneal epithelial-stromal separation [2-fold decrease]). Of the 24 proteins dysregulated upon NM exposure, six proteins (delta-like canonical Notch ligand 1, FGFbasic, CD54, CCL7, endostatin, receptor tyrosine-protein kinase erbB-4) associated with angiogenesis, immune/inflammatory responses, and cell differentiation/proliferation, showed significant reversal upon DEX treatment (Student's t test; P ≤ 0.05). Complementing our animal model studies, DEX was shown to mitigate vesicant-induced toxicities in ex vivo human corneas. SIGNIFICANCE STATEMENT: Nitrogen mustard (NM) exposure-induced injuries were optimized in an ex vivo human cornea culture model and studies were carried out at 24 h post 100 nmoles NM exposure. Dexamethasone (DEX) administration (started 2 h post NM exposure and every 8 h thereafter) reversed NM-induced corneal injuries. Molecular mediators of DEX action were associated with angiogenesis, immune/inflammatory responses, and cell differentiation/proliferation, indicating DEX aids wound healing via reversing vesicant-induced neovascularization (delta-like canonical Notch ligand 1 and FGF basic) and leukocyte infiltration (CD54 and CCL7).
Collapse
Affiliation(s)
- Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - David A Ammar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Neera Tewari-Singh
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Mina B Pantcheva
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - J Mark Petrash
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences (N.M., R.K., K.K., C.A., R.A.) and Department of Ophthalmology, School of Medicine (M.B.P., J.M.P.) University of Colorado-Anschutz Medical Campus, Aurora, Colorado; Lions Eye Institute for Transplant and Research, Tampa, Florida (D.A.A.); and Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan (N.T.-S.)
| |
Collapse
|
11
|
Jalili S, Shirzad H, Mousavi Nezhad SA. Prediction and Validation of Hub Genes Related to Major Depressive Disorder Based on Co-expression Network Analysis. J Mol Neurosci 2024; 74:8. [PMID: 38198075 DOI: 10.1007/s12031-023-02172-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/16/2023] [Indexed: 01/11/2024]
Abstract
Major depressive disorder (MDD) is generally among the most prevalent psychiatric illnesses. Significant advances have occurred in comprehension of the MDD biology. However, it is still essential to recognize new biomarkers for potential targeted treatment of patients with MDD. The present work deals with in-depth comparative computational analyses to obtain new insights, such as gene ontology and pathway enrichment analyses and weighted gene co-expression network analysis (WGCNA) through gene expression dataset. The expression of selected hub-genes was validated in MDD patients using quantitative real-time PCR (RT-qPCR). We found that MDD progression includes the turquoise module genes (p-value = 1e-18, r = 0.97). According to gene enrichment analysis, the cytokine-mediated signaling pathway mostly involves genes in this module. By selection of four candidate hub-genes (IL6, NRG1, TNF, and BDNF), RT-qPCR validation was performed. A significant NRG1 downregulation was revealed by the RT-qPCR outcomes in MDD. In MDD patients, TNF and IL6 expression were considerably higher, and no considerable differences were found in the BDNF expression. Ultimately, based on ROC analyses, IL6, NRG1, and TNF had a higher MDD diagnostic performance. Therefore, our study presents information on the intricate association between MDD development and cytokine-mediated signaling, thus providing new rationales to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Shirin Jalili
- Institute of Police Equipment and Technologies, Policing Sciences and Social Studies Research Institute, Tehran, Iran.
| | - Hadi Shirzad
- Research Center for Life & Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarter, Tehran, Iran.
| | - Seyed Amin Mousavi Nezhad
- Research Center for Life & Health Sciences & Biotechnology of the Police, Directorate of Health, Rescue & Treatment, Police Headquarter, Tehran, Iran
| |
Collapse
|
12
|
Ploypetch S, Wongbandue G, Roytrakul S, Phaonakrop N, Prapaiwan N. Comparative Serum Proteome Profiling of Canine Benign Prostatic Hyperplasia before and after Castration. Animals (Basel) 2023; 13:3853. [PMID: 38136890 PMCID: PMC10740436 DOI: 10.3390/ani13243853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
BPH is the most prevalent prostatic condition in aging dogs. Nevertheless, clinical diagnosis and management remain inconsistent. This study employed in-solution digestion coupled with nano-liquid chromatography tandem mass spectrometry to assess serum proteome profiling of dogs with BPH and those dogs after castration. Male dogs were divided into two groups; control and BPH groups. In the BPH group, each dog was evaluated at two time points: Day 0 (BF subgroup) and Day 30 after castration (AT subgroup). In the BF subgroup, three proteins were significantly upregulated and associated with dihydrotestosterone: solute carrier family 5 member 5, tyrosine-protein kinase, and FRAT regulator of WNT signaling pathway 1. Additionally, the overexpression of polymeric immunoglobulin receptors in the BF subgroup hints at its potential as a novel protein linked to the BPH development process. Conversely, alpha-1-B glycoprotein (A1BG) displayed significant downregulation in the BF subgroup, suggesting A1BG's potential as a predictive protein for canine BPH. Finasteride was associated with increased proteins in the AT subgroup, including apolipoprotein C-I, apolipoprotein E, apolipoprotein A-II, TAO kinase 1, DnaJ homolog subfamily C member 16, PH domain and leucine-rich repeat protein phosphatase 1, neuregulin 1, and pseudopodium enriched atypical kinase 1. In conclusion, this pilot study highlighted alterations in various serum proteins in canine BPH, reflecting different pathological changes occurring in this condition. These proteins could be a source of potential non-invasive biomarkers for diagnosing this disease.
Collapse
Affiliation(s)
- Sekkarin Ploypetch
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| | - Grisnarong Wongbandue
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Narumon Phaonakrop
- Functional Proteomics Technology Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Nawarus Prapaiwan
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand; (S.P.); (G.W.)
| |
Collapse
|
13
|
Eldin AS, Fawzy O, Mahmoud E, Elaziz OHA, Enayet AEA, Khidr EG. Serum neuregulin 1 in relation to ventricular function and subclinical atherosclerosis in type 2 diabetes patients. ENDOCRINOL DIAB NUTR 2023; 70:619-627. [PMID: 38065627 DOI: 10.1016/j.endien.2023.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/26/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND AND AIMS Neuregulin 1 (NRG-1) is one of the members of the epidermal growth factors proteins. The present study provides novel insights into the relationship between serum levels of NRG-1 and insulin resistance, subclinical atherosclerosis and cardiac dysfunction that occur in type 2 diabetes (T2D). METHODS The study included 50 patients with T2D and 40 healthy age- and gender-matched controls. Serum NRG-1 was measured using ELISA. Glycemic parameters, lipid profile and insulin resistance were assessed. Trans-thoracic echocardiography and carotid intima media thickness (CIMT) were studied for all study subjects. RESULTS T2D patients had significantly lower serum NRG-1 levels than controls. Serum NRG-1 was negatively correlated with age, fasting blood glucose, HbA1c, insulin resistance, blood urea, serum creatinine and LDL-C, and positively correlated with HDL-C, eGFR and CIMT. Regarding echocardiographic variables, serum NRG-1 was found to correlate positively with left ventricular global longitudinal strain and negatively with E/Ea ratio. NRG-1 was found to predict subclinical atherosclerosis in type 2 diabetes patients at a cut-off value<108.5pg/ml with 78% sensitivity and 80% specificity. CONCLUSIONS A robust relationship was found between serum NRG-1 levels and hyperglycemia, insulin resistance, subclinical atherosclerosis, and cardiac dysfunction in patients with type 2 diabetes. These results shed light on a possible role of NRG-1 as a potential noninvasive biomarker for detection of cardiometabolic risk in T2D.
Collapse
Affiliation(s)
- Aya Saad Eldin
- Department of Endocrinology and Metabolism, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Olfat Fawzy
- Department of Endocrinology and Metabolism, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Eman Mahmoud
- Department of Endocrinology and Metabolism, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ola H Abd Elaziz
- Department of Cardiology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Alshimaa Ezzat A Enayet
- Department of Radio Diagnosis, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Emad Gamil Khidr
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy for Boys, Al-Azhar University, Nasr City, 11231 Cairo, Egypt.
| |
Collapse
|
14
|
Jiang Y, Shi J, Zhou J, He C, Gu R. ErbB4 promotes M2 activation of macrophages in idiopathic pulmonary fibrosis. Open Life Sci 2023; 18:20220692. [PMID: 37800117 PMCID: PMC10549971 DOI: 10.1515/biol-2022-0692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 10/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and fatal diffuse fibrotic lung disease accompanied by macrophage M2 activation. ErbB4 is involved in and affects the process of inflammation. In this study, we determined that the mRNA level and protein expression of ErbB4 and M2 cytokine members were increased in the serum of IPF patients. In mouse alveolar macrophage MH-S cells, after knocking down ErbB4 by siRNA, the mRNA level and protein expression of M2 activator induced by interleukin (IL)-4 were decreased compared with the control group. Activating by ErbB4 agonist neuromodulatory protein (NRG)-1, IL-4-induced M2 program was promoted. Mechanistically, treated with NRG-1 in MH-S cells, the phosphorylation level of Akt did not change, while the phosphorylation level of ERK increased. Using SCH772984 to inhibit ERK pathway, the increasing IL-4-induced M2 activation by NRG-1 was inhibited, and the high level of M2 activator protein expression and mRNA expression was restored. Collectively, our data support that ErbB4 and M2 programs are implicated in IPF, and ErbB4 participates in the regulation of M2 activation induced by IL-4 through the ERK pathway.
Collapse
Affiliation(s)
- Yu Jiang
- School of Medicine, Shaoxing University, Shaoxing 312000, Zhejiang, China
| | - Jialin Shi
- School of Medicine, Shaoxing University, Shaoxing 312000, Zhejiang, China
| | - Junhao Zhou
- Shaoxing Traditional Chinese Medicine Hospital, Shaoxing 312000, Zhejiang, China
| | - Chunxiao He
- Shaoxing People’s Hospital, Shaoxing 312000, Zhejiang, China
| | - Ruinan Gu
- School of Medicine, Shaoxing University, Shaoxing 312000, Zhejiang, China
| |
Collapse
|
15
|
Sárközy M, Watzinger S, Kovács ZZ, Acar E, Márványkövi F, Szűcs G, Lauber GY, Galla Z, Siska A, Földesi I, Fintha A, Kriston A, Kovács F, Horváth P, Kővári B, Cserni G, Krenács T, Szabó PL, Szabó GT, Monostori P, Zins K, Abraham D, Csont T, Pokreisz P, Podesser BK, Kiss A. Neuregulin-1β Improves Uremic Cardiomyopathy and Renal Dysfunction in Rats. JACC Basic Transl Sci 2023; 8:1160-1176. [PMID: 37791301 PMCID: PMC10543921 DOI: 10.1016/j.jacbts.2023.03.003] [Citation(s) in RCA: 6] [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] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 10/05/2023]
Abstract
Chronic kidney disease is a global health problem affecting 10% to 12% of the population. Uremic cardiomyopathy is often characterized by left ventricular hypertrophy, fibrosis, and diastolic dysfunction. Dysregulation of neuregulin-1β signaling in the heart is a known contributor to heart failure. The systemically administered recombinant human neuregulin-1β for 10 days in our 5/6 nephrectomy-induced model of chronic kidney disease alleviated the progression of uremic cardiomyopathy and kidney dysfunction in type 4 cardiorenal syndrome. The currently presented positive preclinical data warrant clinical studies to confirm the beneficial effects of recombinant human neuregulin-1β in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Simon Watzinger
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Z.A. Kovács
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Eylem Acar
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Fanni Márványkövi
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Gülsüm Yilmaz Lauber
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Zsolt Galla
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Attila Fintha
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
- Single-Cell Technologies Ltd, Szeged, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tibor Krenács
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Gábor Tamás Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Péter Monostori
- Metabolic and Newborn Screening Laboratory, Department of Pediatrics, Albert Szent-Györgyi Medical School, University of Szeged, Hungary
| | - Karin Zins
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Abraham
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Center of Excellence, University of Szeged, Szeged, Hungary
| | - Peter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Yang X, Cheng K, Wang LY, Jiang JG. The role of endothelial cell in cardiac hypertrophy: Focusing on angiogenesis and intercellular crosstalk. Biomed Pharmacother 2023; 163:114799. [PMID: 37121147 DOI: 10.1016/j.biopha.2023.114799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023] Open
Abstract
Cardiac hypertrophy is characterized by cardiac structural remodeling, fibrosis, microvascular rarefaction, and chronic inflammation. The heart is structurally organized by different cell types, including cardiomyocytes, fibroblasts, endothelial cells, and immune cells. These cells highly interact with each other by a number of paracrine or autocrine factors. Cell-cell communication is indispensable for cardiac development, but also plays a vital role in regulating cardiac response to damage. Although cardiomyocytes and fibroblasts are deemed as key regulators of hypertrophic stimulation, other cells, including endothelial cells, also exert important effects on cardiac hypertrophy. More particularly, endothelial cells are the most abundant cells in the heart, which make up the basic structure of blood vessels and are widespread around other cells in the heart, implicating the great and inbuilt advantage of intercellular crosstalk. Cardiac microvascular plexuses are essential for transport of liquids, nutrients, molecules and cells within the heart. Meanwhile, endothelial cell-mediated paracrine signals have multiple positive or negative influences on cardiac hypertrophy. However, a comprehensive discussion of these influences and consequences is required. This review aims to summarize the basic function of endothelial cells in angiogenesis, with an emphasis on angiogenic molecules under hypertrophic conditions. The secondary objective of the research is to fully discuss the key molecules involved in the intercellular crosstalk and the endothelial cell-mediated protective or detrimental effects on other cardiac cells. This review provides a more comprehensive understanding of the overall role of endothelial cells in cardiac hypertrophy and guides the therapeutic approaches and drug development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xing Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China
| | - Kun Cheng
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Lu-Yun Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| | - Jian-Gang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| |
Collapse
|
17
|
Ma Y, Liu Z, Miao L, Jiang X, Ruan H, Xuan R, Xu S. Mechanisms underlying pathological scarring by fibroblasts during wound healing. Int Wound J 2023. [PMID: 36726192 DOI: 10.1111/iwj.14097] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Pathological scarring is an abnormal outcome of wound healing, which often manifests as excessive proliferation and transdifferentiation of fibroblasts (FBs), and excessive deposition of the extracellular matrix. FBs are the most important effector cells involved in wound healing and scar formation. The factors that promote pathological scar formation often act on the proliferation and function of FB. In this study, we describe the factors that lead to abnormal FB formation in pathological scarring in terms of the microenvironment, signalling pathways, epigenetics, and autophagy. These findings suggest that understanding the causes of abnormal FB formation may aid in the development of precise and effective preventive and treatment strategies for pathological scarring that are associated with improved quality of life of patients.
Collapse
Affiliation(s)
- Yizhao Ma
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Zhifang Liu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - LinLin Miao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Xinyu Jiang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Hongyu Ruan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Rongrong Xuan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Suling Xu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
18
|
Zhbanov KA, Salakheeva EY, Sokolova IY, Zheleznykh EA, Zektser VY, Privalova EV, Belenkov YN, Shchendrygina AA. Neuregulin-1β, Biomarkers of Inflammation and Myocardial Fibrosis in Heart Failure Patients. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-09-05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- K. A. Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. Yu. Salakheeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - I. Ya. Sokolova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. A. Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - V. Yu. Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. V. Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Yu. N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | |
Collapse
|
19
|
Mahapatra S, Sharma MVR, Brownson B, Gallicano VE, Gallicano GI. Cardiac inducing colonies halt fibroblast activation and induce cardiac/endothelial cells to move and expand via paracrine signaling. Mol Biol Cell 2022; 33:ar96. [PMID: 35653297 DOI: 10.1091/mbc.e22-02-0032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Myocardial fibrosis (MF), a common event that develops after myocardial infarction, initially is a reparative process but eventually leads to heart failure and sudden cardiac arrest. In MF, the infarct area is replaced by a collagenous-based scar induced by "excessive" collagen deposition from activated cardiac fibroblasts. The scar prevents ventricular wall thinning; however, over time it expands to noninfarcted myocardium. Therapies to prevent fibrosis include reperfusion, anti-fibrotic agents, and ACE inhibitors. Paracrine factor (PF)/stem cell research has recently gained significance as a therapy. We consistently find that cardiac inducing colonies (CiCs) (derived from human germline pluripotent stem cells) secrete PFs at physiologically relevant concentrations that suppress cardiac fibroblast activation and excessive extracellular matrix protein secretion. These factors also affect human cardiomyocytes and endothelial cells by inducing migration/proliferation of both populations into a myocardial wound model. Finally, CiC factors modulate matrix turnover and proinflammation. Taking the results together, we show that CiCs could help tip the balance from fibrosis toward repair.
Collapse
Affiliation(s)
- Samiksha Mahapatra
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| | | | - Breanna Brownson
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Rye High School, Rye, NY 10580
| | - Vaughn E Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145.,Thomas Edison High School, Alexandria, VA 22310
| | - G Ian Gallicano
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20057-145
| |
Collapse
|
20
|
Wang Y, Wei J, Zhang P, Zhang X, Wang Y, Chen W, Zhao Y, Cui X. Neuregulin-1, a potential therapeutic target for cardiac repair. Front Pharmacol 2022; 13:945206. [PMID: 36120374 PMCID: PMC9471952 DOI: 10.3389/fphar.2022.945206] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
NRG1 (Neuregulin-1) is an effective cardiomyocyte proliferator, secreted and released by endothelial vascular cells, and affects the cardiovascular system. It plays a major role in heart growth, proliferation, differentiation, apoptosis, and other cardiovascular processes. Numerous experiments have shown that NRG1 can repair the heart in the pathophysiology of atherosclerosis, myocardial infarction, ischemia reperfusion, heart failure, cardiomyopathy and other cardiovascular diseases. NRG1 can connect related signaling pathways through the NRG1/ErbB pathway, which form signal cascades to improve the myocardial microenvironment, such as regulating cardiac inflammation, oxidative stress, necrotic apoptosis. Here, we summarize recent research advances on the molecular mechanisms of NRG1, elucidate the contribution of NRG1 to cardiovascular disease, discuss therapeutic approaches targeting NRG1 associated with cardiovascular disease, and highlight areas for future research.
Collapse
Affiliation(s)
- Yan Wang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianliang Wei
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Peng Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yifei Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjing Chen
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanan Zhao
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| | - Xiangning Cui
- Department of Cardiovascular, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanan Zhao, ; Xiangning Cui,
| |
Collapse
|
21
|
Banerjee S, Mishra S, Xu W, Thompson WE, Chowdhury I. Neuregulin-1 signaling regulates cytokines and chemokines expression and secretion in granulosa cell. J Ovarian Res 2022; 15:86. [PMID: 35883098 PMCID: PMC9316729 DOI: 10.1186/s13048-022-01021-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Granulosa cells (GCs) are multilayered somatic cells within the follicle that provide physical support and microenvironment for the developing oocyte. In recent years, the role of Neuregulin-1 (NRG1), a member of the EGF-like factor family, has received considerable attention due to its neurodevelopmental and cardiac function. However, the exact physiological role of NRG1 in GC is mainly unknown. In order to confirm that NRG1 plays a regulatory role in rat GC functions, endogenous NRG1-knockdown studies were carried out in GCs using RNA interference methodology. RESULTS Knockdown of NRG1 in GCs resulted in the enhanced expression and secretion of the cytokines and chemokines. In addition, the phosphorylation of PI3K/Akt/ERK1/2 was significantly low in GCs under these experimental conditions. Moreover, in vitro experimental studies suggest that tumor necrosis factor-α (TNFα) treatment causes the physical destruction of GCs by activating caspase-3/7 activity. In contrast, exogenous NRG1 co-treatment of GCs delayed the onset of TNFα-induced apoptosis and inhibited the activation of caspase-3/7 activity. Furthermore, current experimental studies suggest that gonadotropins promote differential expression of NRG1 and ErbB3 receptors in GCs of the antral follicle. Interestingly, NRG1 and ErbB3 were intensely co-localized in the mural and cumulus GCs and cumulus-oocyte complex of pre-ovulatory follicles in the estrus stage. CONCLUSIONS The present studies suggest that gonadotropins-dependent NRG1-signaling in GCs may require the balance of the cytokines and chemokines expression and secretion, ultimately which may be supporting the follicular maturation and oocyte competence for ovulation and preventing follicular atresia.
Collapse
Affiliation(s)
- Saswati Banerjee
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Sameer Mishra
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive Southwest, Atlanta, GA, 30310, USA
| | - Wei Xu
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Winston E Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Indrajit Chowdhury
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive Southwest, Atlanta, GA, 30310, USA.
| |
Collapse
|
22
|
Prophylactic Evidence of MSCs-Derived Exosomes in Doxorubicin/Trastuzumab-Induced Cardiotoxicity: Beyond Mechanistic Target of NRG-1/Erb Signaling Pathway. Int J Mol Sci 2022; 23:ijms23115967. [PMID: 35682646 PMCID: PMC9181089 DOI: 10.3390/ijms23115967] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/06/2022] Open
Abstract
Trastuzumab (Trz) is a humanized monoclonal antibody targeting epidermal growth factor receptor 2 (HER2; ErbB2). The combined administration of Trz and doxorubicin (DOX) has shown potent anti-cancer efficacy; however, this regimen may be accompanied by severe cardiac toxicity. Mesenchymal stem cells (MSCs)-derived exosomes are nanosized vesicles that play a crucial role in cell–cell communication and have shown efficacy in the treatment of various diseases. In this study, we aim to investigate the cardioprotective effects of MSCs-derived exosomes in a DOX/Trz- mediated cardiotoxicity model, and the possible mechanisms underlying these effects are elucidated. Forty-nine male rats were randomly assigned into four groups: Group I (control); Group II (Dox/Trz); Group III (protective group); and Group IV (curative group). Cardiac hemodynamic parameters, serum markers of cardiac injury, oxidative stress indices, and cardiac histopathology were investigated. Further, transcript profile of specific cardiac tissue injury markers, apoptotic markers, and fibrotic markers were analyzed using qRT-PCR, while the protein expressions of pAkt/Akt, pERK/ERK, pJNK/JNK, pJNK/JNK, and pSTAT3/STAT3 were evaluated by ELISA. Additionally, cardiac mirR-21 and miR-26a were assessed. A combined administration of DOX/Trz disrupted redox and Ca2+ homeostasis in cardiac tissue induced myocardial fibrosis and myofibril loss and triggered cardiac DNA damage and apoptosis. This cardiotoxicity was accompanied by decreased NRG-1 mRNA expression, HER2 protein expression, and suppressed AKT and ERK phosphorylation, while triggering JNK phosphorylation. Histological and ultra-structural examination of cardiac specimens revealed features typical of cardiac tissue injury. Moreover, a significant decline in cardiac function was observed through biochemical testing of serum cardiac markers and echocardiography. In contrast, the intraperitoneal administration of MSCs-derived exosomes alleviated cardiac injury in both protective and curative protocols; however, superior effects were observed in the protective protocol. The results of the current study indicate the ability of MSCs-derived exosomes to protect from and attenuate DOX/Trz-induced cardiotoxicity. The NRG-1/HER2, MAPK, PI3K/AKT, PJNK/JNK, and PSTAT/STAT signaling pathways play roles in mediating these effects.
Collapse
|
23
|
Rocco E, Grimaldi MC, Maino A, Cappannoli L, Pedicino D, Liuzzo G, Biasucci LM. Advances and Challenges in Biomarkers Use for Coronary Microvascular Dysfunction: From Bench to Clinical Practice. J Clin Med 2022; 11:2055. [PMID: 35407662 PMCID: PMC8999821 DOI: 10.3390/jcm11072055] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 02/01/2023] Open
Abstract
Coronary microvascular dysfunction (CMD) is related to a broad variety of clinical scenarios in which cardiac microvasculature is morphologically and functionally affected, and it is associated with impaired responses to vasoactive stimuli. Although the prevalence of CMD involves about half of all patients with chronic coronary syndromes and more than 20% of those with acute coronary syndrome, the diagnosis of CMD is often missed, leading to the underestimation of its clinical importance. The established and validated techniques for the measurement of coronary microvascular function are invasive and expensive. An ideal method to assess endothelial dysfunction should be accurate, non-invasive, cost-effective and accessible. There are varieties of biomarkers available, potentially involved in microvascular disease, but none have been extensively validated in this heterogeneous clinical population. The investigation of potential biomarkers linked to microvascular dysfunction might improve the assessment of the diagnosis, risk stratification, disease progression and therapy response. This review article offers an update about traditional and novel potential biomarkers linked to CMD.
Collapse
Affiliation(s)
- Erica Rocco
- Department of Medical-Surgical Sciences and Biotechnologies, Cardiology Unit, ICOT Hospital, Sapienza University of Rome, 04110 Latina, Italy;
| | - Maria Chiara Grimaldi
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandro Maino
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
| | - Luigi Cappannoli
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
| | - Daniela Pedicino
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Luigi Marzio Biasucci
- Department of Cardiovascular and Pneumological Sciences, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.M.); (L.C.); (D.P.); (G.L.); (L.M.B.)
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
24
|
Shiraishi M, Yamaguchi A, Suzuki K. Nrg1/ErbB signaling-mediated regulation of fibrosis after myocardial infarction. FASEB J 2022; 36:e22150. [PMID: 34997943 DOI: 10.1096/fj.202101428rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/11/2022]
Abstract
Appropriate fibrotic tissue formation after myocardial infarction (MI) is crucial to the maintenance of the heart's structure. M2-like macrophages play a vital role in post-MI fibrosis by activating cardiac fibroblasts. Because the mechanism by which post-MI cardiac fibrosis is regulated is not fully understood, we investigated, in vitro and in vivo, the cellular and molecular mechanisms of post-MI fibrotic tissue formation, especially those related to the regulation of cellular senescence and apoptosis. CD206+ F4/80+ CD11b+ M2-like macrophages collected from mouse hearts on post-MI day 7 showed increased expression of neuregulin 1 (Nrg1). Nrg1 receptor epidermal growth factor receptors ErbB2 and ErbB4 were expressed on cardiac fibroblasts in the infarct area. M2-like macrophage-derived Nrg1 suppressed both hydrogen peroxide-induced senescence and apoptosis of fibroblasts, whereas blockade of ErbB function significantly accelerated both processes. M2-like macrophage-derived Nrg1/ErbB/PI3K/Akt signaling, shown to be related to anti-senescence, was activated in damaged cardiac fibroblasts. Interestingly, systemic blockade of ErbB function in MI model mice enhanced senescence and apoptosis of cardiac fibroblasts and exacerbated inflammation. Further, increased accumulation of M2-like macrophages resulted in excessive post-MI progression of fibrosis in mice hearts. The molecular mechanism underlying the regulation of fibrotic tissue formation in the infarcted myocardium was shown in part to be attenuation of apoptosis and senescence of cardiac fibroblasts by the activation of Nrg1/ErbB/PI3K/Akt signaling. M2-like macrophage-mediated regulation of Nrg1/ErbB signaling has a substantial effect on fibrotic tissue formation in the infarcted adult mouse heart and is critical for suppressing the progression of senescence and apoptosis of cardiac fibroblasts.
Collapse
Affiliation(s)
- Manabu Shiraishi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan.,William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Atsushi Yamaguchi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
25
|
Tang S, Xiao G, Yuan Q, Lin W, Yuan X, Fang X, Deng T, Xiao X. Long Non-coding RNA ENST00000453774.1 Confers an Inhibitory Effect on Renal Fibrosis by Inhibiting miR-324-3p to Promote NRG1 Expression. Front Cell Dev Biol 2021; 9:580754. [PMID: 34869303 PMCID: PMC8640469 DOI: 10.3389/fcell.2021.580754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 04/08/2021] [Indexed: 01/07/2023] Open
Abstract
Progressive or chronic renal diseases arise from a process of destructive renal fibrosis. Therefore, the molecular basis of renal fibrosis has attracted increasing attention. In this investigation, we set out to elucidate the potential interaction among long non-coding RNA ENST00000453774.1 (lncRNA 74.1), microRNA-324-3p (miR-324-3p), and NRG1, and to investigate their roles in the context of cellular autophagy and renal fibrosis. We collected 30 renal fibrosis tissue samples for analysis. In other studies, HK-2 cells were stimulated with TGF-β1 to induce a cell model of renal fibrosis, followed by alteration on the expression of lncRNA 74.1, miR-324-3p, or NRG1, or by the addition of AKT activator SC79 in the HK-2 cells. The expression levels of lncRNA 74.1, miR-324-3p, NRG1, autophagy-related proteins (ATG5, ATG7, LC3II/I, and P62), and the corresponding fibrosis markers (Collagen I, Fibronectin, and α-SMA) were subsequently determined using various assay methods. In addition, the proportion of LC3 positive cells and number of autophagosomes were recorded. Results revealed that lncRNA 74.1 and NRG1 were poorly expressed and miR-324-3p was highly expressed in renal fibrosis tissues and modeled cells. LncRNA 74.1 could bind to miR-324-3p, which led to upregulated NRG1 expression and inhibition of the PI3K/AKT signaling pathway. Meanwhile, overexpression of lncRNA 74.1 or down-regulation of miR-324-3p increased the levels of ATG5, ATG7, LC3II, and LC3I, and decreased levels of P62, Collagen I, Fibronectin, and α-SMA, accompanied by elevated proportions of LC3 positive cells and autophagosomes. Findings concur in showing that lncRNA 74.1 could induce cellular autophagy and alleviate renal fibrosis by regulating the miR-324-3p-mediated NRG1/PI3K/AKT axis. This axis may thus present a potential molecular target in renal fibrosis treatment.
Collapse
Affiliation(s)
- Shumei Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Gong Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Qiongjing Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangning Yuan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Fang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Tianci Deng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Veeder JA, Hothem LN, Cipriani AE, Jensen BC, Rodgers JE. Chemotherapy-associated cardiomyopathy: Mechanisms of toxicity and cardioprotective strategies. Pharmacotherapy 2021; 41:1066-1080. [PMID: 34806206 DOI: 10.1002/phar.2638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To describe the proposed mechanisms of chemotherapy-associated cardiomyopathy (CAC) and potential cardioprotective therapies for CAC including a comprehensive review of existing systematic analyses, guideline recommendations, and ongoing clinical trials. DATA SOURCES A literature search of MEDLINE was performed (from 1990 to June 2020) using the following search terms: anthracycline, trastuzumab, cardiomyopathy, cardiotoxicity, primary prevention, angiotensin-converting enzyme inhibitor (ACEI), angiotensin receptor blocker (ARB), beta blocker, dexrazoxane (DEX) as well as using individual names from select therapeutic categories. STUDY SELECTION AND DATA EXTRACTION Existing English language systematic analyses and guidelines were considered. DATA SYNTHESIS The mechanisms of CAC are multifaceted, but various cardioprotective therapies target many of these pathways. To date, anthracyclines and HER-2 targeted therapies have been the focus of cardioprotective trials to date as they are the most commonly implicated therapies in CAC. While traditional neurohormonal antagonists (ACEIs, ARBs, and beta blockers) and DEX performed favorably in many small clinical trials, the quality of available evidence remains limited. Hence, major guidelines lack consensus on an approach to primary prevention of CAC. Given the uncertain role of preventive therapy, monitoring for a symptomatic or asymptomatic decline in LV function is imperative with prompt evaluation should this occur. Numerous ongoing randomized controlled trials seek to either confirm the findings of these previous studies or identify new therapeutic agents to prevent CAC. Clinical implications are derived from the available literature as well as current guideline recommendations for CAC cardioprotection. CONCLUSION At this time, no single therapy has a clear cardioprotective benefit in preventing CAC nor is any therapy strongly recommended by current guidelines. Additional studies are needed to determine the optimal preventative regimens.
Collapse
Affiliation(s)
- Justin A Veeder
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- AstraZeneca, Nashville, Tennessee, USA
| | - Lauren N Hothem
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- GlaxoSmithKline, Research Triangle, North Carolina, USA
| | - Amber E Cipriani
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, North Carolina, USA
| | - Brian C Jensen
- Department of Medicine, University of North Carolina Medical Center, Chapel Hill, North Carolina, USA
| | - Jo E Rodgers
- UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
| |
Collapse
|
27
|
Conte E. Targeting monocytes/macrophages in fibrosis and cancer diseases: Therapeutic approaches. Pharmacol Ther 2021; 234:108031. [PMID: 34774879 DOI: 10.1016/j.pharmthera.2021.108031] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023]
Abstract
Over almost 140 years since their identification, the knowledge about macrophages has unbelievably evolved. The 'big eaters' from being thought of as simple phagocytic cells have been recognized as master regulators in immunity, homeostasis, healing/repair and organ development. Long considered to originate exclusively from bone marrow-derived circulating monocytes, macrophages have been also demonstrated to be the first immune cells colonizing tissues in the developing embryo and persisting in adult life by self-renewal, as long-lived tissue resident macrophages. Therefore, heterogeneous populations of macrophages with different ontogeny and functions co-exist in tissues. Macrophages act as sentinels of homeostasis and are intrinsically programmed to lead the wound healing and repair processes that occur after injury. However, in certain pathological circumstances macrophages get dysfunctional, and impaired or aberrant macrophage activities become key features of diseases. For instance, in both fibrosis and cancer, that have been defined 'wounds that do not heal', dysfunctional monocyte-derived macrophages overall play a key detrimental role. On the other hand, due to their plasticity these cells can be 're-educated' and exert anti-fibrotic and anti-cancer functions. Therefore macrophages represent an important therapeutic target in both fibrosis and cancer diseases. The current review will illustrate new insights into the role of monocytes/macrophages in these devastating diseases and summarize new therapeutic strategies and applications of macrophage-targeted drug development in their clinical setting.
Collapse
|
28
|
Hu X, Xu H, Xue Q, Wen R, Jiao W, Tian K. The role of ERBB4 mutations in the prognosis of advanced non-small cell lung cancer treated with immune checkpoint inhibitors. Mol Med 2021; 27:126. [PMID: 34620079 PMCID: PMC8496027 DOI: 10.1186/s10020-021-00387-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have witnessed the achievements of convincing clinical benefits that feature the significantly prolonged overall survival (OS) of patients suffering from advanced non-small cell lung cancer (NSCLC), according to reports recently. Sensitivity to immunotherapy is related to several biomarkers, such as PD-L1 expression, TMB level, MSI-H and MMR. However, a further investigation into the novel biomarkers of the prognosis on ICIs treatment is required. In addition, there is an urgent demand for the establishment of a systematic hazard model to assess the efficacy of ICIs therapy for advanced NSCLC patients. METHODS In this study, the gene mutation and clinical data of NSCLC patients was obtained from the TCGA database, followed by the analysis of the detailed clinical information and mutational data relating to two advanced NSCLC cohorts receiving the ICIs treatment from the cBioPortal of Cancer Genomics. The Kaplan-Meier plot method was used to perform survival analyses, while selected variables were adopted to develop a systematic nomogram. The prognostic significance of ERBB4 in pan-cancer was analyzed by another cohort from the cBioPortal of Cancer Genomics. RESULTS The mutation frequencies of TP53 and ERBB4 were 54% and 8% in NSCLC, respectively. The mutual exclusive analysis in cBioPortal has indicated that ERBB4 does show co-occurencing mutations with TP53. Patients with ERBB4 mutations were confirmed to have better prognosis for ICIs treatment, compared to those seeing ERBB4 wild type (PFS: exact p = 0.017; OS: exact p < 0.01) and only TP53 mutations (OS: p = 0.021). The mutation status of ERBB4 and TP53 was tightly linked to DCB of ICIs treatment, PD-L1 expression, TMB value, and TIICs. Finally, a novel nomogram was built to evaluate the efficacy of ICIs therapy. CONCLUSION ERBB4 mutations could serve as a predictive biomarker for the prognosis of ICIs treatment. The systematic nomogram was proven to have the great potential for evaluating the efficacy of ICIs therapy for advanced NSCLC patients.
Collapse
Affiliation(s)
- Xilin Hu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Hanlin Xu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Qianwen Xue
- Qingdao Maternal & Child Health and Family Planning Service Center, Qingdao, 266000, Shandong, China
| | - Ruran Wen
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Kaihua Tian
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
29
|
Kishore A, Petrek M. Roles of Macrophage Polarization and Macrophage-Derived miRNAs in Pulmonary Fibrosis. Front Immunol 2021; 12:678457. [PMID: 34489932 PMCID: PMC8417529 DOI: 10.3389/fimmu.2021.678457] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
This mini-review summarizes the current evidence for the role of macrophage activation and polarization in inflammation and immune response pertinent to interstitial lung disease, specifically pulmonary fibrosis. In the fibrosing lung, the production and function of inflammatory and fibrogenic mediators involved in the disease development have been reported to be regulated by the effects of polarized M1/M2 macrophage populations. The M1 and M2 macrophage phenotypes were suggested to correspond with the pro-inflammatory and pro-fibrogenic signatures, respectively. These responses towards tissue injury followed by the development and progression of lung fibrosis are further regulated by macrophage-derived microRNAs (miRNAs). Besides cellular miRNAs, extracellular exosomal-miRNAs derived from M2 macrophages have also been proposed to promote the progression of pulmonary fibrosis. In a future perspective, harnessing the noncoding miRNAs with a key role in the macrophage polarization is, therefore, suggested as a promising therapeutic strategy for this debilitating disease.
Collapse
Affiliation(s)
- Amit Kishore
- Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Accuscript Consultancy, Ludhiana, India
| | - Martin Petrek
- Department of Pathological Physiology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czechia.,Departments of Experimental Medicine, and Immunology, University Hospital Olomouc, Olomouc, Czechia
| |
Collapse
|
30
|
Dang UJ, Ziemba M, Clemens PR, Hathout Y, Conklin LS, Hoffman EP. Serum biomarkers associated with baseline clinical severity in young steroid-naïve Duchenne muscular dystrophy boys. Hum Mol Genet 2021; 29:2481-2495. [PMID: 32592467 PMCID: PMC7471506 DOI: 10.1093/hmg/ddaa132] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/28/2020] [Accepted: 06/20/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin in muscle, and while all patients share the primary gene and biochemical defect, there is considerable patient–patient variability in clinical symptoms. We sought to develop multivariate models of serum protein biomarkers that explained observed variation, using functional outcome measures as proxies for severity. Serum samples from 39 steroid-naïve DMD boys 4 to <7 years enrolled into a clinical trial of vamorolone were studied (NCT02760264). Four assessments of gross motor function were carried out for each participant over a 6-week interval, and their mean was used as response for biomarker models. Weighted correlation network analysis was used for unsupervised clustering of 1305 proteins quantified using SOMAscan® aptamer profiling to define highly representative and connected proteins. Multivariate models of biomarkers were obtained for time to stand performance (strength phenotype; 17 proteins) and 6 min walk performance (endurance phenotype; 17 proteins) including some shared proteins. Identified proteins were tested with associations of mRNA expression with histological severity of muscle from dystrophinopathy patients (n = 28) and normal controls (n = 6). Strong associations predictive of both clinical and histological severity were found for ERBB4 (reductions in both blood and muscle with increasing severity), SOD1 (reductions in muscle and increases in blood with increasing severity) and CNTF (decreased levels in blood and muscle with increasing severity). We show that performance of DMD boys was effectively modeled with serum proteins, proximal strength associated with growth and remodeling pathways and muscle endurance centered on TGFβ and fibrosis pathways in muscle.
Collapse
Affiliation(s)
- Utkarsh J Dang
- Department of Health Outcomes and Administrative Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Michael Ziemba
- Department of Biomedical Engineering, Watson School of Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Paula R Clemens
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Veteran Affairs Medical Center, Pittsburgh, PA 15213, USA
| | - Yetrib Hathout
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | | | | | - Eric P Hoffman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA.,ReveraGen BioPharma, Rockville, MD 20850, USA
| |
Collapse
|
31
|
Feyen E, Ricke-Hoch M, Van Fraeyenhove J, Vermeulen Z, Scherr M, Dugaucquier L, Viereck J, Bruyns T, Thum T, Segers VFM, Hilfiker-Kleiner D, De Keulenaer GW. ERBB4 and Multiple MicroRNAs That Target ERBB4 Participate in Pregnancy-Related Cardiomyopathy. Circ Heart Fail 2021; 14:e006898. [PMID: 34247489 DOI: 10.1161/circheartfailure.120.006898] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Peripartum cardiomyopathy (PPCM) is a life-threatening disease in women without previously known cardiovascular disease. It is characterized by a sudden onset of heart failure before or after delivery. Previous studies revealed that the generation of a 16-kDa PRL (prolactin) metabolite, the subsequent upregulation of miR-146a, and the downregulation of the target gene Erbb4 is a common driving factor of PPCM. METHODS miRNA profiling was performed in plasma of PPCM patients (n=33) and postpartum-matched healthy CTRLs (controls; n=36). Elevated miRNAs in PPCM plasma, potentially targeting ERBB4 (erythroblastic leukemia viral oncogene homolog 4), were overexpressed in cardiomyocytes using lentiviral vectors. Next, cardiac function, cardiac morphology, and PPCM phenotype were investigated after recurrent pregnancies of HZ (heterozygous) cardiomyocyte-specific Erbb4 mice (Erbb4F/+ αMHC-Cre+, n=9) with their age-matched nonpregnant CTRLs (n=9-10). RESULTS Here, we identify 9 additional highly conserved miRNAs (miR-199a-5p and miR-199a-3p, miR-145a-5p, miR-130a-3p, miR-135a-5p, miR-221-3p, miR-222-3p, miR-23a-3p, and miR19b-3p) that target tyrosine kinase receptor ERBB4 and are over 4-fold upregulated in plasma of PPCM patients at the time of diagnosis. We confirmed that miR-146a, miR-199a-5p, miR-221-3p, miR-222-3p, miR-23a-3p, miR-130a-5p, and miR-135-3p overexpression decreases ERBB4 expression in cardiomyocytes (-29% to -50%; P<0.05). In addition, we demonstrate that genetic cardiomyocyte-specific downregulation of Erbb4 during pregnancy suffices to induce a variant of PPCM in mice, characterized by left ventricular dilatation (postpartum second delivery: left ventricular internal diameter in diastole, +19±7% versus HZ-CTRL; P<0.05), increased atrial natriuretic peptide (ANP) levels (4-fold increase versus HZ-CTRL mice, P<0.001), decreased VEGF (vascular endothelial growth factor) and VE-cadherin levels (-33±17%, P=0.07; -27±20%, P<0.05 versus HZ-CTRL), and histologically enlarged cardiomyocytes (+20±21%, versus HZ-CTRL, P<0.05) but without signs of myocardial apoptosis and inflammation. CONCLUSIONS ERBB4 is essential to protect the maternal heart from peripartum stress. Downregulation of ERBB4 in cardiomyocytes induced by multiple miRNAs in the peripartum period may be crucial in PPCM pathophysiology. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT00998556.
Collapse
Affiliation(s)
- Eline Feyen
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology (M.R.-H., D.H.-K.), Hannover Medical School, Germany
| | - Jens Van Fraeyenhove
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| | - Zarha Vermeulen
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation (M.S.), Hannover Medical School, Germany
| | - Lindsey Dugaucquier
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| | - Janika Viereck
- Institute of Molecular and Translational Therapeutic Strategies (J.V., T.T.), Hannover Medical School, Germany
| | - Tine Bruyns
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (J.V., T.T.), Hannover Medical School, Germany
| | - Vincent F M Segers
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.).,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium (V.F.M.S.). Department of Cardiology, ZNA Hospital, Antwerp, Belgium (G.W.D.K.)
| | | | - Gilles W De Keulenaer
- Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium (E.F., J.V.f., Z.V., L.D., T.B., V.F.M.S., G.W.D.K.)
| |
Collapse
|
32
|
Tsou PS, Palisoc PJ, Ali M, Khanna D, Sawalha AH. Genome-Wide Reduction in Chromatin Accessibility and Unique Transcription Factor Footprints in Endothelial Cells and Fibroblasts in Scleroderma Skin. Arthritis Rheumatol 2021; 73:1501-1513. [PMID: 33586346 DOI: 10.1002/art.41694] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/22/2020] [Accepted: 02/09/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is characterized by widespread fibrosis and vascular complications. This study was undertaken to examine the chromatin landscape and transcription factor footprints in SSc, using an assay for genome-wide chromatin accessibility. METHODS Dermal endothelial cells (ECs) and fibroblasts were isolated from healthy controls and patients with diffuse cutaneous SSc (dcSSc). Assay for transposase-accessible chromatin with sequencing (ATAC-seq) was performed to assess genome-wide chromatin accessibility at a read depth of ~150 million reads per sample. Transcription factor footprinting and motif binding analysis were performed, followed by functional experiments. RESULTS Chromatin accessibility was significantly reduced in dcSSc patients compared to healthy controls. Differentially accessible chromatin loci were enriched in pathways and gene ontologies involved in the nervous system, cell membrane projections and cilia motility, nuclear and steroid receptors, and nitric oxide. In addition, chromatin binding of transcription factors SNAI2, ETV2, and ELF1 was significantly increased in dcSSc ECs, while recruitment of RUNX1 and RUNX2 was enriched in dcSSc fibroblasts. We found significant down-regulation of the neuronal gene NRXN1 and up-regulation of SNAI2 and ETV2 in dcSSc ECs. In dcSSc fibroblasts, down-regulation of the neuronal gene ENTPD1 and up-regulation of RUNX2 were confirmed. Further functional analysis revealed that ETV2 and NRXN1 dysregulation affected angiogenesis in ECs, while ENTPD1 enhanced profibrotic properties in dcSSc fibroblasts. CONCLUSION Our data identify the chromatin blueprint of dcSSc, and suggest that neuronal-related characteristics of SSc ECs and fibroblasts could be a culprit for dysregulated angiogenesis and enhanced fibrosis. Targeting the key pathways and transcription factors identified might present novel therapeutic approaches in SSc.
Collapse
|
33
|
Nishat S, Gumina RJ. The chicken, the egg, and the elephant: eNOS and NRG1 in fibrosis. Am J Physiol Heart Circ Physiol 2021; 321:H292-H293. [PMID: 34170198 DOI: 10.1152/ajpheart.00308.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shamama Nishat
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Department of Cellular Biology and Physiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Richard J Gumina
- The Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Department of Cellular Biology and Physiology, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
34
|
Shen YH, Abe JI. Nrg1β as a Proangiogenesis Therapy: How Is Nrg1β Therapy Unique From Other Angiogenesis Therapies? Arterioscler Thromb Vasc Biol 2021; 41:2315-2317. [PMID: 34162231 DOI: 10.1161/atvbaha.121.316513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ying H Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, TX (Y.H.S.)
| | - Jun-Ichi Abe
- Division of Internal Medicine, Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston (J.I.A.)
| |
Collapse
|
35
|
Schumacher MA, Dennis IC, Liu CY, Robinson C, Shang J, Bernard JK, Washington MK, Polk DB, Frey MR. NRG4-ErbB4 signaling represses proinflammatory macrophage activity. Am J Physiol Gastrointest Liver Physiol 2021; 320:G990-G1001. [PMID: 33826403 PMCID: PMC8285586 DOI: 10.1152/ajpgi.00296.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 03/11/2021] [Accepted: 04/01/2021] [Indexed: 01/31/2023]
Abstract
Proinflammatory macrophages are essential drivers of colitis and express the growth factor receptor ErbB4. This study tested the role of ErbB4 and its specific ligand, NRG4, in regulating macrophage function. We show that endogenous NRG4-ErbB4 signaling limits macrophage production of proinflammatory cytokines in vitro and limits colitis severity in vivo and thus is a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Michael A Schumacher
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Isabella C Dennis
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Cambrian Y Liu
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Cache Robinson
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Judie Shang
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Jessica K Bernard
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - D Brent Polk
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
36
|
Shakeri H, Boen JRA, De Moudt S, Hendrickx JO, Leloup AJA, Jacobs G, De Meyer GRY, De Keulenaer GW, Guns PJDF, Segers VFM. Neuregulin-1 compensates for endothelial nitric oxide synthase deficiency. Am J Physiol Heart Circ Physiol 2021; 320:H2416-H2428. [PMID: 33989083 DOI: 10.1152/ajpheart.00914.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Endothelial cells (ECs) secrete different paracrine signals that modulate the function of adjacent cells; two examples of these paracrine signals are nitric oxide (NO) and neuregulin-1 (NRG1), a cardioprotective growth factor. Currently, it is undetermined whether one paracrine factor can compensate for the loss of another. Herein, we hypothesized that NRG1 can compensate for endothelial NO synthase (eNOS) deficiency. We characterized eNOS null and wild-type (WT) mice by cardiac ultrasound and histology and we determined circulating NRG1 levels. In a separate experiment, eight groups of mice were divided into four groups of eNOS null mice and WT mice; half of the mice received angiotensin II (ANG II) to induce a more severe phenotype. Mice were randomized to daily injections with NRG1 or vehicle for 28 days. eNOS deficiency increased NRG1 plasma levels, indicating that ECs increase their NRG1 expression when NO production is deleted. eNOS deficiency also increased blood pressure, lowered heart rate, induced cardiac fibrosis, and affected diastolic function. In eNOS null mice, ANG II administration not only increased cardiac fibrosis but also induced cardiac hypertrophy and renal fibrosis. NRG1 administration prevented cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. Moreover, Nrg1 expression in the myocardium is shown to be regulated by miR-134. This study indicates that administration of endothelium-derived NRG1 can compensate for eNOS deficiency in the heart and kidneys.NEW & NOTEWORTHY ECs compensate for eNOS deficiency by increasing the secretion of NRG1. NRG1 administration prevents cardiac and renal hypertrophy and fibrosis caused by ANG II infusion and eNOS deficiency. NRG1 expression is regulated by miR-134.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jente R A Boen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Antwerp, Belgium
| | - Sofie De Moudt
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jhana O Hendrickx
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Arthur J A Leloup
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Griet Jacobs
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Hartcentrum ZNA, Antwerp, Belgium
| | | | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
37
|
Neuregulins: protective and reparative growth factors in multiple forms of cardiovascular disease. Clin Sci (Lond) 2021; 134:2623-2643. [PMID: 33063822 PMCID: PMC7557502 DOI: 10.1042/cs20200230] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Neuregulins (NRGs) are protein ligands that act through ErbB receptor tyrosine kinases to regulate tissue morphogenesis, plasticity, and adaptive responses to physiologic needs in multiple tissues, including the heart and circulatory system. The role of NRG/ErbB signaling in cardiovascular biology, and how it responds to physiologic and pathologic stresses is a rapidly evolving field. While initial concepts focused on the role that NRG may play in regulating cardiac myocyte responses, including cell survival, growth, adaptation to stress, and proliferation, emerging data support a broader role for NRGs in the regulation of metabolism, inflammation, and fibrosis in response to injury. The constellation of effects modulated by NRGs may account for the findings that two distinct forms of recombinant NRG-1 have beneficial effects on cardiac function in humans with systolic heart failure. NRG-4 has recently emerged as an adipokine with similar potential to regulate cardiovascular responses to inflammation and injury. Beyond systolic heart failure, NRGs appear to have beneficial effects in diastolic heart failure, prevention of atherosclerosis, preventing adverse effects on diabetes on the heart and vasculature, including atherosclerosis, as well as the cardiac dysfunction associated with sepsis. Collectively, this literature supports the further examination of how this developmentally critical signaling system functions and how it might be leveraged to treat cardiovascular disease.
Collapse
|
38
|
Yu X, Fang C, Liu L, Zhao X, Liu W, Cao H, Lv S. Transcriptome study underling difference of milk yield during peak lactation of Kazakh horse. J Equine Vet Sci 2021; 102:103424. [PMID: 34119198 DOI: 10.1016/j.jevs.2021.103424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/15/2021] [Accepted: 02/15/2021] [Indexed: 01/10/2023]
Abstract
This study was designed to provide a basis for further understanding of the mechanism of lactation based on mRNA expression differences in milk fat between different milk yields in Kazakh horses. Total RNA was extracted from the milk fat during the peak of lactation period. A total of 310 differentially expressed genes (DEGs) were identified by comparative transcriptome analysis of the high-yield and low-yield group. These DEGs regulate lactation by participated in AMPK signaling pathway, FoxO signaling pathway, ErbB signaling pathway, VEGF signaling pathway. In addition, we performed quantitative PCR to validated 5 selected DEGs and the results were in agreement with RNA-seq analysis. A new profile has been established for revealing the mechanism of equid's mammalian lactation.
Collapse
Affiliation(s)
- Xi Yu
- Xinjiang Agricultural University, Urumuqi, China
| | | | - Lingling Liu
- Xinjiang Agricultural University, Urumuqi, China
| | | | - Wujun Liu
- Xinjiang Agricultural University, Urumuqi, China.
| | - Hang Cao
- Xinjiang Agricultural University, Urumuqi, China
| | - Shipeng Lv
- Xinjiang Agricultural University, Urumuqi, China
| |
Collapse
|
39
|
Pathomechanisms and therapeutic opportunities in radiation-induced heart disease: from bench to bedside. Clin Res Cardiol 2021; 110:507-531. [PMID: 33591377 PMCID: PMC8055626 DOI: 10.1007/s00392-021-01809-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/16/2021] [Indexed: 12/14/2022]
Abstract
Cancer management has undergone significant improvements, which led to increased long-term survival rates among cancer patients. Radiotherapy (RT) has an important role in the treatment of thoracic tumors, including breast, lung, and esophageal cancer, or Hodgkin's lymphoma. RT aims to kill tumor cells; however, it may have deleterious side effects on the surrounding normal tissues. The syndrome of unwanted cardiovascular adverse effects of thoracic RT is termed radiation-induced heart disease (RIHD), and the risk of developing RIHD is a critical concern in current oncology practice. Premature ischemic heart disease, cardiomyopathy, heart failure, valve abnormalities, and electrical conduct defects are common forms of RIHD. The underlying mechanisms of RIHD are still not entirely clear, and specific therapeutic interventions are missing. In this review, we focus on the molecular pathomechanisms of acute and chronic RIHD and propose preventive measures and possible pharmacological strategies to minimize the burden of RIHD.
Collapse
|
40
|
Abstract
The myocardium consists of different cell types, of which endothelial cells, cardiomyocytes, and fibroblasts are the most abundant. Communication between these different cell types, also called paracrine signaling, is essential for normal cardiac function, but also important in cardiac remodeling and heart failure. Systematic studies on the expression of ligands and their corresponding receptors in different cell types showed that for 60% of the expressed ligands in a particular cell, the receptor is also expressed. The fact that many ligand-receptor pairs are present in most cells, including the major cell types in the heart, indicates that autocrine signaling is a widespread phenomenon. Autocrine signaling in cardiac remodeling and heart failure is involved in all pathophysiological mechanisms generally observed: hypertrophy, fibrosis, angiogenesis, cell survival, and inflammation. Herein, we review ligand-receptor pairs present in the major cardiac cell types based on RNA-sequencing expression databases, and we review current literature on extracellular signaling proteins with an autocrine function in the heart; these include C-type natriuretic peptide, fibroblast growth factors 2, F21, and 23, macrophage migration inhibitory factor, heparin binding-epidermal growth factor, angiopoietin-like protein 2, leptin, adiponectin, follistatin-like 1, apelin, neuregulin 1, vascular endothelial growth factor, transforming growth factor β, wingless-type integration site family, member 1-induced secreted protein-1, interleukin 11, connective tissue growth factor/cellular communication network factor, and calcitonin gene‒related peptide. The large number of autocrine signaling factors that have been studied in the literature supports the concept that autocrine signaling is an essential part of myocardial biology and disease.
Collapse
Affiliation(s)
- Vincent F. M. Segers
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyUniversity Hospital AntwerpEdegemBelgium
| | - Gilles W. De Keulenaer
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyZNA HospitalAntwerpBelgium
| |
Collapse
|
41
|
Screening for potential targets to reduce stenosis in bioprosthetic heart valves. Sci Rep 2021; 11:2464. [PMID: 33510256 PMCID: PMC7843970 DOI: 10.1038/s41598-021-81340-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/24/2020] [Indexed: 12/02/2022] Open
Abstract
Progressive stenosis is one of the main factors that limit the lifetime of bioprosthetic valved conduits. To improve long-term performance we aimed to identify targets that inhibit pannus formation on conduit walls. From 11 explanted, obstructed, RNAlater presevered pulmonary valved conduits, we dissected the thickened conduit wall and the thin leaflet to determine gene expression-profiles using ultra deep sequencing. Differential gene expression between pannus and leaflet provided the dataset that was screened for potential targets. Promising target candidates were immunohistologically stained to see protein abundance and the expressing cell type(s). While immunostainings for DDR2 and FGFR2 remained inconclusive, EGFR, ErbB4 and FLT4 were specifically expressed in a subset of tissue macrophages, a cell type known to regulate the initiation, maintenance, and resolution of tissue repair. Taken toghether, our data suggest EGFR, ErbB4 and FLT4 as potential target candidates to limit pannus formation in bioprosthestic replacement valves.
Collapse
|
42
|
Shchendrigina AA, Zhbanov KA, Privalova EV, Iusupova AO, Bytdaeva AH, Danilogorskaya YA, Zheleznykh EA, Suvorov AY, Zektser VY, Mnatsakanyan MG, Lyapidevskaya OV, Khabarova NV, Naymann YI, Belenkov YN, Starostina ES. [Circulating Neuregulin-1 and Chronic Heart Failure with Preserved Ejection]. ACTA ACUST UNITED AC 2020; 60:1222. [PMID: 33487159 DOI: 10.18087/cardio.2020.11.n1222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/08/2020] [Indexed: 11/18/2022]
Abstract
Chronic heart failure (CHF) with preserved ejection fraction (CHFpEF) is an unsolved, socially relevant challenge since it is associated with a high level of morbidity and mortality. Early markers for this pathology are unavailable, and therapeutic approaches are undeveloped. This necessitates extensive studying the mechanisms of CHFpEF to identify therapeutic targets. According to current notions, systemic inflammation and endothelial dysfunction play an important role in the pathogenesis of CHFpEF. These processes induce the development of myocardial fibrosis and impairment of cardiomyocyte relaxation, thereby resulting in diastolic dysfunction and increased left ventricular (LV) filling pressure. Neuregulin-1 (NRG-1) is a paracrine growth factor and a natural agonist of ErbB receptor family synthesized in the endothelium of coronary microvessels. The NRG-1 / ErbB4 system of the heart is activated at early stages of CHFpEF to enhance the cardiomyocyte resistance to oxidative stress. Preclinical and clinical (phases II and III) studies have shown that the recombinant NRG-1 therapy results in improvement of myocardial contractility and in LV reverse remodeling. Results of recent studies suggest possible anti-inflammatory and antifibrotic effects of NRG-1, which warrants studying the activity of this system in patients with CHFpEF.
Collapse
Affiliation(s)
- A A Shchendrigina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - K A Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E V Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A O Iusupova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A H Bytdaeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu A Danilogorskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E A Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | | - V Yu Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - M G Mnatsakanyan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - O V Lyapidevskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - N V Khabarova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu I Naymann
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu N Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | |
Collapse
|
43
|
Yin H, Favreau-Lessard AJ, deKay JT, Herrmann YR, Robich MP, Koza RA, Prudovsky I, Sawyer DB, Ryzhov S. Protective role of ErbB3 signaling in myeloid cells during adaptation to cardiac pressure overload. J Mol Cell Cardiol 2020; 152:1-16. [PMID: 33259856 DOI: 10.1016/j.yjmcc.2020.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/06/2020] [Accepted: 11/23/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Myeloid cells play an important role in a wide variety of cardiovascular disorders, including both ischemic and non-ischemic cardiomyopathies. Neuregulin-1 (NRG-1)/ErbB signaling has recently emerged as an important factor contributing to the control of inflammatory activation of myeloid cells after an ischemic injury. However, the role of ErbB signaling in myeloid cells in non-ischemic cardiomyopathy is not fully understood. This study investigated the role of ErbB3 receptors in the regulation of early adaptive response using a mouse model of transverse aortic constriction (TAC) for non-ischemic cardiomyopathy. METHODS AND RESULTS TAC surgery was performed in groups of age- and sex-matched myeloid cell-specific ErbB3-deficient mice (ErbB3MyeKO) and control animals (ErbB3MyeWT). The number of cardiac CD45 immune cells, CD11b myeloid cells, Ly6G neutrophils, and Ly6C monocytes was determined using flow cytometric analysis. Five days after TAC, survival was dramatically reduced in male but not female ErbB3MyeKO mice or control animals. The examination of lung weight to body weight ratio suggested that acute pulmonary edema was present in ErbB3MyeKO male mice after TAC. To determine the cellular and molecular mechanisms involved in the increased mortality in ErbB3MyeKO male mice, cardiac cell populations were examined at day 3 post-TAC using flow cytometry. Myeloid cells accumulated in control but not in ErbB3MyeKO male mouse hearts. This was accompanied by increased proliferation of Sca-1 positive non-immune cells (endothelial cells and fibroblasts) in control but not ErbB3MyeKO male mice. No significant differences in intramyocardial accumulation of myeloid cells or proliferation of Sca-1 cells were found between the groups of ErbB3MyeKO and ErbB3MyeWT female mice. An antibody-based protein array analysis revealed that IGF-1 expression was significantly downregulated only in ErbB3MyeKO mice hearts compared to control animals after TAC. CONCLUSION Our data demonstrate the crucial role of myeloid cell-specific ErbB3 signaling in the cardiac accumulation of myeloid cells, which contributes to the activation of cardiac endothelial cells and fibroblasts and development of an early adaptive response to cardiac pressure overload in male mice.
Collapse
Affiliation(s)
- Haifeng Yin
- Maine Medical Center Research Institute, Scarborough, ME, United States of America
| | | | - Joanne T deKay
- Maine Medical Center Research Institute, Scarborough, ME, United States of America
| | - Yodit R Herrmann
- Maine Medical Center Research Institute, Scarborough, ME, United States of America
| | - Michael P Robich
- Maine Medical Center Research Institute, Scarborough, ME, United States of America; Maine Medical Center, Cardiovascular Institute, Portland, ME, United States of America
| | - Robert A Koza
- Maine Medical Center Research Institute, Scarborough, ME, United States of America
| | - Igor Prudovsky
- Maine Medical Center Research Institute, Scarborough, ME, United States of America
| | - Douglas B Sawyer
- Maine Medical Center Research Institute, Scarborough, ME, United States of America; Maine Medical Center, Cardiovascular Institute, Portland, ME, United States of America
| | - Sergey Ryzhov
- Maine Medical Center Research Institute, Scarborough, ME, United States of America.
| |
Collapse
|
44
|
Triposkiadis F, Butler J, Abboud FM, Armstrong PW, Adamopoulos S, Atherton JJ, Backs J, Bauersachs J, Burkhoff D, Bonow RO, Chopra VK, de Boer RA, de Windt L, Hamdani N, Hasenfuss G, Heymans S, Hulot JS, Konstam M, Lee RT, Linke WA, Lunde IG, Lyon AR, Maack C, Mann DL, Mebazaa A, Mentz RJ, Nihoyannopoulos P, Papp Z, Parissis J, Pedrazzini T, Rosano G, Rouleau J, Seferovic PM, Shah AM, Starling RC, Tocchetti CG, Trochu JN, Thum T, Zannad F, Brutsaert DL, Segers VF, De Keulenaer GW. The continuous heart failure spectrum: moving beyond an ejection fraction classification. Eur Heart J 2020; 40:2155-2163. [PMID: 30957868 DOI: 10.1093/eurheartj/ehz158] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/05/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022] Open
Abstract
Randomized clinical trials initially used heart failure (HF) patients with low left ventricular ejection fraction (LVEF) to select study populations with high risk to enhance statistical power. However, this use of LVEF in clinical trials has led to oversimplification of the scientific view of a complex syndrome. Descriptive terms such as 'HFrEF' (HF with reduced LVEF), 'HFpEF' (HF with preserved LVEF), and more recently 'HFmrEF' (HF with mid-range LVEF), assigned on arbitrary LVEF cut-off points, have gradually arisen as separate diseases, implying distinct pathophysiologies. In this article, based on pathophysiological reasoning, we challenge the paradigm of classifying HF according to LVEF. Instead, we propose that HF is a heterogeneous syndrome in which disease progression is associated with a dynamic evolution of functional and structural changes leading to unique disease trajectories creating a spectrum of phenotypes with overlapping and distinct characteristics. Moreover, we argue that by recognizing the spectral nature of the disease a novel stratification will arise from new technologies and scientific insights that will shape the design of future trials based on deeper understanding beyond the LVEF construct alone.
Collapse
Affiliation(s)
| | - Javed Butler
- Department of Medicine-L650, University of Mississippi Medical Center, Jackson, MS, USA
| | - Francois M Abboud
- Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| | - Paul W Armstrong
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Stamatis Adamopoulos
- Transplant and Mechanical Circulatory Support Unit, Onassis Cardiac Surgery Center, Athens, Greece
| | - John J Atherton
- Department of Cardiology, Royal Brisbane and Women's Hospital, University of Queensland School of Medicine, Brisbane, Australia
| | - Johannes Backs
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Heidelberg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | | | - Robert O Bonow
- Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Northwestern Memorial Hospital, Chicago, IL, USA
| | - Vijay K Chopra
- Department of Cardiology, Medanta Medicity, Gurugram, Haryana, India
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Leon de Windt
- Department of Cardiology, Faculty of Health, Medicine and Life Sciences, School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University Bochum, Bochum, Germany
| | - Gerd Hasenfuss
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Jean-Sébastien Hulot
- Université Paris-Descartes, Sorbonne Paris Cité, Paris, France.,Paris Cardiovascular Research Center, INSERM UMR 970, Paris, France.,Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Marvin Konstam
- The CardioVascular Center of Tufts Medical Center, Boston, MA, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Wolfgang A Linke
- Institute of Physiology II, University of Münster, Münster, Germany
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Alexander R Lyon
- Cardiovascular Research Centre, Royal Brompton Hospital, London, UK.,National Heart and Lung Institute, Imperial College London, London, UK
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Douglas L Mann
- Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis Missouri, MO, USA
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care Medicine, AP-HP, Saint Louis and Lariboisière University Hospitals, Inserm U 942, Paris, France
| | | | | | - Zoltan Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - John Parissis
- Heart Failure Unit, Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Department of Cardiovascular Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | - Giuseppe Rosano
- Department of Medical Sciences, IRCCS San Raffaele, Centre for Clinical and Basic Research, Pisana Rome, Italy
| | - Jean Rouleau
- Montreal Heart Institute and University of Montreal, Montreal, Quebec, Canada
| | | | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre, King's College London, London, UK
| | | | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Jean-Noel Trochu
- CIC INSERM 1413, Institut du thorax, UMR INSERM 1087, University Hospital of Nantes, Nantes, France
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hanover, Germany
| | - Faiez Zannad
- Inserm CIC 1433, Université de Lorrain, CHU de Nancy, Nancy, France
| | | | - Vincent F Segers
- Laboratory of Physiopharmacology, Antwerp University, Universiteitsplein 1, Building T, Wilrijk, Antwerp, Belgium.,Division of Cardiology, Antwerp University Hospital, Edegem, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, Antwerp University, Universiteitsplein 1, Building T, Wilrijk, Antwerp, Belgium.,ZNA Hartcentrum, Antwerp, Belgium
| |
Collapse
|
45
|
Zhu Z, Xu X, Wang F, Song Y, Zhu Y, Quan W, Zhang X, Bi C, He H, Li S, Li X. Integrative microRNA and mRNA expression profiling in acute aristolochic acid nephropathy in mice. Mol Med Rep 2020; 22:3367-3377. [PMID: 32945497 PMCID: PMC7453650 DOI: 10.3892/mmr.2020.11444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
In acute aristolochic acid nephropathy (AAN), aristolochic acid (AA) induces renal injury and tubulointerstitial fibrosis. However, the roles of microRNAs (miRNAs/miRs) and mRNAs involved in AAN are not clearly understood. The aim of the present study was to examine AA‑induced genome‑wide differentially expressed (DE) miRNAs and DE mRNAs using deep sequencing in mouse kidneys, and to analyze their regulatory networks. In the present self‑controlled study, mice were treated with 5 mg/kg/day AA for 5 days, following unilateral nephrectomy. AA‑induced renal injury and tubulointerstitial fibrosis were detected using hematoxylin and eosin staining and Masson's trichrome staining in the mouse kidneys. A total of 82 DE miRNAs and 4,605 DE mRNAs were identified between the AA‑treated group and the self‑control group. Of these DE miRNAs and mRNAs, some were validated using reverse transcription‑quantitative PCR. Expression levels of the profibrotic miR‑21, miR‑433 and miR‑132 families were significantly increased, whereas expression levels of the anti‑fibrotic miR‑122‑5p and let‑7a‑1‑3p were significantly decreased. Functions and signaling pathways associated with the DE miRNAs and mRNAs were analyzed using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG). A total of 767 DE pairs (in opposing directions) of miRNAs and their mRNA targets were identified. Among these, regulatory networks of miRNAs and mRNAs were analyzed using KEGG to identify enriched signaling pathways and extracellular matrix‑associated pathways. In conclusion, the present study identified genome‑wide DE miRNAs and mRNAs in the kidneys of AA‑treated mice, as well as their regulatory pairs and signaling networks. The present results may improve the understanding of the role of DE miRNAs and their mRNA targets in the pathophysiology of acute AAN.
Collapse
Affiliation(s)
- Ziqiang Zhu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xinxing Xu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Fengying Wang
- Department of Pediatrics, Sir Run Hospital, Nanjing Medical University, Nanjing, Jiangsu 211100, P.R. China
| | - Yongrui Song
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yanping Zhu
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wei Quan
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xueli Zhang
- Centre for Systems Biology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
- School of Medicine, Institute of Medical Sciences, Örebro University, SE-70182 Örebro, Sweden
| | - Cheng Bi
- Centre for Systems Biology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Hongxin He
- Centre for Systems Biology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shuang Li
- Centre for Systems Biology, Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiaozhong Li
- Department of Nephrology and Immunology, Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
46
|
Baci D, Bosi A, Parisi L, Buono G, Mortara L, Ambrosio G, Bruno A. Innate Immunity Effector Cells as Inflammatory Drivers of Cardiac Fibrosis. Int J Mol Sci 2020; 21:E7165. [PMID: 32998408 PMCID: PMC7583949 DOI: 10.3390/ijms21197165] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Despite relevant advances made in therapies for cardiovascular diseases (CVDs), they still represent the first cause of death worldwide. Cardiac fibrosis and excessive extracellular matrix (ECM) remodeling are common end-organ features in diseased hearts, leading to tissue stiffness, impaired myocardial functional, and progression to heart failure. Although fibrosis has been largely recognized to accompany and complicate various CVDs, events and mechanisms driving and governing fibrosis are still not entirely elucidated, and clinical interventions targeting cardiac fibrosis are not yet available. Immune cell types, both from innate and adaptive immunity, are involved not just in the classical response to pathogens, but they take an active part in "sterile" inflammation, in response to ischemia and other forms of injury. In this context, different cell types infiltrate the injured heart and release distinct pro-inflammatory cytokines that initiate the fibrotic response by triggering myofibroblast activation. The complex interplay between immune cells, fibroblasts, and other non-immune/host-derived cells is now considered as the major driving force of cardiac fibrosis. Here, we review and discuss the contribution of inflammatory cells of innate immunity, including neutrophils, macrophages, natural killer cells, eosinophils and mast cells, in modulating the myocardial microenvironment, by orchestrating the fibrogenic process in response to tissue injury. A better understanding of the time frame, sequences of events during immune cells infiltration, and their action in the injured inflammatory heart environment, may provide a rationale to design new and more efficacious therapeutic interventions to reduce cardiac fibrosis.
Collapse
Affiliation(s)
- Denisa Baci
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Annalisa Bosi
- Laboratory of Pharmacology, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy;
| | - Luca Parisi
- Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, University of Milan, 20122 Milan, Italy;
| | - Giuseppe Buono
- Unit of Immunology, IRCCS MultiMedica, 20138 Milan, Italy;
| | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Giuseppe Ambrosio
- Division of Cardiology, University of Perugia School of Medicine, 06123 Perugia, Italy;
| | - Antonino Bruno
- Unit of Immunology, IRCCS MultiMedica, 20138 Milan, Italy;
| |
Collapse
|
47
|
Dugaucquier L, Feyen E, Mateiu L, Bruyns TAM, De Keulenaer GW, Segers VFM. The role of endothelial autocrine NRG1/ERBB4 signaling in cardiac remodeling. Am J Physiol Heart Circ Physiol 2020; 319:H443-H455. [PMID: 32618511 DOI: 10.1152/ajpheart.00176.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuregulin-1 (NRG1) is a paracrine growth factor, secreted by cardiac endothelial cells (ECs) in conditions of cardiac overload/injury. The current concept is that the cardiac effects of NRG1 are mediated by activation of erythroblastic leukemia viral oncogene homolog (ERBB)4/ERBB2 receptors on cardiomyocytes. However, recent studies have shown that paracrine effects of NRG1 on fibroblasts and macrophages are equally important. Here, we hypothesize that NRG1 autocrine signaling plays a role in cardiac remodeling. We generated EC-specific Erbb4 knockout mice to eliminate endothelial autocrine ERBB4 signaling without affecting paracrine NRG1/ERBB4 signaling in the heart. We first observed no basal cardiac phenotype in these mice up to 32 wk. We next studied these mice following transverse aortic constriction (TAC), exposure to angiotensin II (ANG II), or myocardial infarction in terms of cardiac performance, myocardial hypertrophy, myocardial fibrosis, and capillary density. In general, no major differences between EC-specific Erbb4 knockout mice and control littermates were observed. However, 8 wk following TAC both myocardial hypertrophy and fibrosis were attenuated by EC-specific Erbb4 deletion, albeit these responses were normalized after 20 wk. Similarly, 4 wk after ANG II treatment, myocardial fibrosis was less pronounced compared with control littermates. These observations were supported by RNA-sequencing experiments on cultured endothelial cells showing that NRG1 controls the expression of various hypertrophic and fibrotic pathways. Overall, this study shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling. This study contributes to understanding the spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury.NEW & NOTEWORTHY The role of NRG1/ERBB signaling in endothelial cells is not completely understood. Our study contributes to the understanding of spatiotemporal heterogeneity of myocardial autocrine and paracrine responses following cardiac injury and shows a role of endothelial autocrine NRG1/ERBB4 signaling in the modulation of hypertrophic and fibrotic responses during early cardiac remodeling.
Collapse
Affiliation(s)
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | | | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, Middelheim Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
48
|
Amani M, Rahmati M, Fathi M, Ahmadvand H. Reduce Muscle Fibrosis through Exercise via NRG1/ErbB2 Modification in Diabetic Rats. J Diabetes Res 2020; 2020:6053161. [PMID: 32509881 PMCID: PMC7244949 DOI: 10.1155/2020/6053161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/02/2019] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic myopathy refers to the manifestations in the skeletal muscle as a result of altered glucose homeostasis which reflects as fibrosis. Since physical exercise has been indicated a protective strategy for improving glucose metabolism in skeletal muscle, we tested a hypothesis under which the endurance exercise training could reverse the produced skeletal muscle fibrosis by diabetes. Eight-week-old male Wistar rats were randomly assigned into four groups including healthy control (HC), healthy trained (HT), diabetic control (DC), and diabetic trained (DT) groups. Diabetes was induced by a single intraperitoneal injection of streptozotocin (STZ; 45 mg/kg). Rats in the HT and DT groups carried out an exercise program on a motorized treadmill for five days a week over six weeks. Skeletal muscle levels of NRG1and ErbB2 were measured by the Western blot method. Exercise training decreased blood glucose levels in the DT group. Induction of diabetes increased skeletal muscle fibrosis in both the fast extensor digitorum longus (EDL) and slow soleus muscles, while endurance training modified it in diabetic trained rats. Moreover, muscle NRG1and ErbB2 levels were increased in diabetic rats, while training modified muscle NRG1and ErbB2 levels in diabetic trained rats. Our study provides novel evidence that endurance training could modify skeletal muscle fibrosis through NRG1/ErbB2 modification in STZ-induced diabetic rats.
Collapse
MESH Headings
- Animals
- Diabetes Complications/metabolism
- Diabetes Complications/prevention & control
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/therapy
- Fibrosis/metabolism
- Fibrosis/prevention & control
- Male
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Atrophy/metabolism
- Muscular Atrophy/prevention & control
- Neuregulin-1/metabolism
- Physical Conditioning, Animal/physiology
- Rats
- Rats, Wistar
- Receptor, ErbB-2/metabolism
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Majid Amani
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| | - Masoud Rahmati
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| | - Mohammad Fathi
- Department of Physical Education and Sport Sciences, Faculty of Literature and Human Sciences, Lorestan University, Khoramabad, Iran
| | - Hasan Ahmadvand
- Department of Biochemistry, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
49
|
Hage C, Wärdell E, Linde C, Donal E, Lam CS, Daubert C, Lund LH, Månsson‐Broberg A. Circulating neuregulin1-β in heart failure with preserved and reduced left ventricular ejection fraction. ESC Heart Fail 2020; 7:445-455. [PMID: 31981321 PMCID: PMC7160501 DOI: 10.1002/ehf2.12615] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/22/2019] [Accepted: 12/23/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Neuregulin1-β (NRG1-β) is released from microvascular endothelial cells in response to inflammation with compensatory cardioprotective effects. Circulating NRG1-β is elevated in heart failure (HF) with reduced ejection fraction (HFrEF) but not studied in HF with preserved EF (HFpEF). METHODS AND RESULTS Circulating NRG1-β was quantified in 86 stable patients with HFpEF (EF ≥45% and N-terminal pro-brain natriuretic peptide >300 ng/L), in 86 patients with HFrEF prior to and after left ventricular assist device (LVAD) and/or heart transplantation (HTx) and in 21 healthy controls. Association between NRG1-β and the composite outcome of all-cause mortality/HF hospitalization in HFpEF and all-cause mortality/HTx/LVAD implantation in HFrEF with and without ischaemia assessed as macrovascular coronary artery disease was assessed. In HFpEF, median (25th-75th percentile) NRG1-β was 6.5 (2.1-11.3) ng/mL; in HFrEF, 3.6 (2.1-7.6) ng/mL (P = 0.035); after LVAD, 1.7 (0.9-3.6) ng/mL; after HTx 2.1 (1.4-3.6) ng/mL (overall P < 0.001); and in controls, 29.0 (23.1-34.3) ng/mL (P = 0.001). In HFrEF, higher NRG1-β was associated with worse outcomes (hazard ratio per log increase 1.45, 95% confidence interval 1.04-2.03, P = 0.029), regardless of ischaemia. In HFpEF, the association of NRG1-β with outcomes was modified by ischaemia (log-rank P = 0.020; Pinteraction = 0.553) such that only in ischaemic patients, higher NRG1-β was related to worse outcomes. In contrast, in patients without ischaemia, higher NRG1-β trended towards better outcomes (hazard ratio 0.71, 95% confidence interval 0.48-1.05, P = 0.085). CONCLUSIONS Neuregulin1-β was reduced in HFpEF and further reduced in HFrEF. The opposing relationships of NRG1-β with outcomes in non-ischaemic HFpEF compared with HFrEF and ischaemic HFpEF may indicate compensatory increases of cardioprotective NRG1-β from microvascular endothelial dysfunction in the former (non-ischaemic HFpEF), but this compensatory mechanism is overwhelmed by the presence of ischaemia in the latter (HFrEF and ischaemic HFpEF).
Collapse
Affiliation(s)
- Camilla Hage
- Department of MedicineKarolinska Institutet – SolnaStockholmSweden
- Heart and Vascular ThemeKarolinska University HospitalSE‐171 76StockholmSweden
| | - Eva Wärdell
- Department of MedicineKarolinska Institutet – HuddingeHuddingeSweden
- Heart and Vascular ThemeKarolinska University HospitalSE‐171 76StockholmSweden
| | - Cecilia Linde
- Department of MedicineKarolinska Institutet – SolnaStockholmSweden
- Heart and Vascular ThemeKarolinska University HospitalSE‐171 76StockholmSweden
| | - Erwan Donal
- Département de Cardiologie & CIC‐IT U 804Centre Hospitalier Universitaire de RennesRennesFrance
| | - Carolyn S.P. Lam
- National Heart Centre SingaporeDuke‐National University of SingaporeSingaporeSingapore
- University Medical Centre GroningenGroningenThe Netherlands
- The George Institute for Global HealthSydneyAustralia
| | - Claude Daubert
- Département de Cardiologie & CIC‐IT U 804Centre Hospitalier Universitaire de RennesRennesFrance
| | - Lars H. Lund
- Department of MedicineKarolinska Institutet – SolnaStockholmSweden
- Heart and Vascular ThemeKarolinska University HospitalSE‐171 76StockholmSweden
| | - Agneta Månsson‐Broberg
- Department of MedicineKarolinska Institutet – HuddingeHuddingeSweden
- Heart and Vascular ThemeKarolinska University HospitalSE‐171 76StockholmSweden
| |
Collapse
|
50
|
Segers VFM, Dugaucquier L, Feyen E, Shakeri H, De Keulenaer GW. The role of ErbB4 in cancer. Cell Oncol (Dordr) 2020; 43:335-352. [PMID: 32219702 DOI: 10.1007/s13402-020-00499-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The epidermal growth factor receptor family consists of four members, ErbB1 (epidermal growth factor receptor-1), ErbB2, ErbB3, and ErbB4, which all have been found to play important roles in tumor development. ErbB4 appears to be unique among these receptors, because it is the only member with growth inhibiting properties. ErbB4 plays well-defined roles in normal tissue development, in particular the heart, the nervous system, and the mammary gland system. In recent years, information on the role of ErbB4 in a number of tumors has emerged and its general direction points towards a tumor suppressor role for ErbB4. However, there are some controversies and conflicting data, warranting a review on this topic. CONCLUSIONS Here, we discuss the role of ErbB4 in normal physiology and in breast, lung, colorectal, gastric, pancreatic, prostate, bladder, and brain cancers, as well as in hepatocellular carcinoma, cholangiocarcinoma, and melanoma. Understanding the role of ErbB4 in cancer is not only important for the treatment of tumors, but also for the treatment of other disorders in which ErbB4 plays a major role, e.g. cardiovascular disease.
Collapse
Affiliation(s)
- Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium. .,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium.
| | - Lindsey Dugaucquier
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Eline Feyen
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.,Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| |
Collapse
|