1
|
Güvenç M, Aydin T, Kutlu T, Etyemez M, İşler CT. Tomentosin mitigates the LPS induced cardiac injury by regulating Nrf-2/Nf-κβ pathway in mice. Eur J Pharmacol 2025; 996:177589. [PMID: 40187600 DOI: 10.1016/j.ejphar.2025.177589] [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: 10/28/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Endotoxemic shock is a severe complication characterized by multiple organ failure, hypotension, and impaired tissue perfusion, all contributing to high morbidity and mortality. Recent studies have highlighted the anti-inflammatory and antioxidant properties of tomentosin. This study investigates the protective effects of tomentosin against lipopolysaccharide (LPS)-induced cardiac injury and elucidates its underlying mechanisms. Mice were pre-treated with tomentosin before the LPS administration. Subsequently, cardiac injury markers, oxidative stress parameters, inflammatory mediators, and Nrf-2/NF-κB protein expression levels were analysed. The results demonstrated that tomentosin significantly reduced Troponin and CK-MB levels, alleviated oxidative stress, and suppressed inflammatory responses. Furthermore, tomentosin inhibited NF-κB activation while enhancing Nrf-2 expression. In conclusion, our findings suggest that tomentosin exerts cardioprotective effects by modulating the Nrf-2/NF-κB pathway, positioning it as a potential therapeutic candidate for preventing LPS-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Mehmet Güvenç
- Department of Physiology, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey.
| | - Tuba Aydin
- Department of Pharmacognosy, Faculty of Pharmacy, Ağrı İbrahim Çeçen University, Ağrı, 04100, Turkey
| | - Tuncer Kutlu
- Department of Pathology, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey
| | - Muhammed Etyemez
- Department of Physiology, Faculty of Veterinary Medicine, University of Kastamonu, Kastamonu, 37150, Turkey
| | - Cafer Tayer İşler
- Department of Surgery, Faculty of Veterinary Medicine, University of Hatay Mustafa Kemal, Hatay, 31060, Turkey
| |
Collapse
|
2
|
Maj M, Hernik K, Tyszkiewicz K, Owe-Larsson M, Sztokfisz-Ignasiak A, Malejczyk J, Janiuk I. A complex role of chromogranin A and its peptides in inflammation, autoimmunity, and infections. Front Immunol 2025; 16:1567874. [PMID: 40370467 PMCID: PMC12074958 DOI: 10.3389/fimmu.2025.1567874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025] Open
Abstract
Chromogranin A (CgA), mostly known as a nonspecific neuroendocrine tumor marker, was the first glycoprotein from the granin family characterized as a prohormone for various bioactive peptides including vasostatin I/II (VS-I, VS-II), catestatin (CST), chromofungin (CHR), pancreastatin (PST), WE-14, and others. CgA and its derivatives present various functions, often antagonistic, in maintaining body homeostasis and influencing the immune system. This review aims to summarize the not fully understood role of CgA and its derivatives in inflammation, autoimmunity, and infections. CgA seems to be involved in the complex pathophysiology of cardiovascular disorders, neurodegenerative diseases, and other conditions where immune system dysfunction plays a role in the onset and development of the disease (e.g. systemic lupus erythematosus (SLE), inflammatory bowel disease (IBD), or rheumatoid arthritis (RA)). However, the direct immunomodulatory role of CgA is difficult to assess since many of its activities may be linked with its peptides. CST and VS-I are considered anti-inflammatory molecules, due to M2 macrophage polarization stimulation and downregulation of certain proinflammatory cytokines. Conversely, PST is reported to stimulate proinflammatory M1 macrophage polarization and Th1 lymphocyte response. Thus, the final effects of CgA in inflammation may depend on its cleavage pattern. Additionally, peptides like CST, VS-I, or CHR exert direct antimicrobial/antifungal activities. CgA, WE-14, and other less-known CgA-derived peptides have also been reported to trigger autoimmune responses, highly studied in type 1 diabetes mellitus. Overall, CgA and its derivatives have an interesting but complex role in immunity, however, their specific roles require further research.
Collapse
Affiliation(s)
- Maciej Maj
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Karolina Hernik
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Kaja Tyszkiewicz
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Maja Owe-Larsson
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Alicja Sztokfisz-Ignasiak
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Jacek Malejczyk
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
- Institute of Health Sciences, Faculty of Medical and Health Sciences, University of Siedlce, Siedlce, Poland
| | - Izabela Janiuk
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
De Bartolo A, Romeo N, Marrone A, Rago V, Granieri MC, Vommaro ML, Cupelli A, Cerra MC, Indiveri C, Ronca R, Cantile M, Sanna R, Rocca C, Angelone T. A recombinant fragment antigen-binding (Fab) of trastuzumab displays low cytotoxic profile in adult human cardiomyocytes: first evidence and the key implication of FcγRIIA receptor. Acta Pharmacol Sin 2025; 46:618-631. [PMID: 39414958 PMCID: PMC11845480 DOI: 10.1038/s41401-024-01397-3] [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: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
Fragment crystallizable gamma receptors (FcγRs) mediate various cellular responses with significant cardiovascular implications. They contribute to the anticancer activity of trastuzumab (TRZ), a recombinant humanized monoclonal antibody that interferes with human epidermal growth factor receptor 2 (HER2), thereby blocking its physiological function in cardiac cells. This is responsible for cardiac complications that hamper TRZ clinical application. In this study we investigated the involvement of FcγRs in the TRZ cardiotoxicity. We used a recombinant antigen-binding fragment (Fab) of TRZ (rFab-HER2) to examine whether the absence of the Fc region resulted in fewer cardiomyocyte toxicity while preserving TRZ's ability to inhibit HER2. When exposed to rFab-HER2, AC16 human adult ventricular cardiomyocytes were less vulnerable to damage and death, than to TRZ. Specifically, TRZ exhibited cytotoxicity at a lower concentration (150 µg/mL, corresponding to ~1 µM) compared to rFab-HER2 (250 µg/mL, corresponding to ~5 µM). Like TRZ, rFab-HER2 negatively modulated HER2 levels in cardiomyocyte (without inducing cytotoxic activity in BJ human fibroblast cells that either did not express or express very low levels of HER2) and inhibited the downstream ERK/AKT cascades. But rFab-HER2 did not alter cardiomyocyte mitochondrial dynamic balance, and affect apoptosis and inflammation, while it limited cytosolic and mitochondrial ROS indicators. On contrary, the Fc region (50-250 μg/mL) exerted direct cytotoxic action on cardiomyocytes (but not on human fibroblasts that lacked Fc receptors). TRZ (150 μg/mL) markedly upregulated the expression level of FcγRIIA (a FcγRs strongly involved in TRZ-induced antibody-dependent cellular toxicity) in cardiomyocytes, whereas the Fab fragment (150 μg/mL) had no effect. Our results demonstrate that Fc region plays an important pathogenic role in TRZ-induced cardiomyocyte toxicity. In addition, targeting FcγRIIA might contribute to the off-target effects of TRZ therapy.
Collapse
Affiliation(s)
- Anna De Bartolo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Naomi Romeo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Alessandro Marrone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| | - Maria Concetta Granieri
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Luigia Vommaro
- Department of Biology, E. and E. S. (DiBEST), University of Calabria, Rende, Italy
| | - Arianna Cupelli
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Carmela Cerra
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Cesare Indiveri
- Department of Biology, E. and E. S. (DiBEST), Unit of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, Cosenza, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| | | | | | | | - Carmine Rocca
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Tommaso Angelone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
4
|
Wu X, Li L, Jinhabure, Xiaofeng, Eerdunchaolu. Radix Sophorae Flavescentis of Sophora flavescens Aiton inhibits LPS-induced macrophage pro-inflammatory response via regulating CFHR2 expression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118210. [PMID: 38641074 DOI: 10.1016/j.jep.2024.118210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Long-term chronic inflammation often leads to chronic diseases. Although Sophora flavescens has been shown to have anti-inflammatory properties, its detailed molecular mechanism is still unknown. AIM OF STUDY This study investigated the effect of Radix Sophorae Flavescentis on the LPS-induced inflammatory response in macrophages. MATERIALS AND METHODS LPS was used to induce the peritoneal macrophages to simulate the inflammatory environment in vitro. Different concentrations of Radix Sophorae Flavescentis-containing (medicated) serum were used for intervention. The peritoneal macrophages were identified by using hematoxylin-eosin and immunofluorescence staining. ELISA was used to measure the TNF-α and IL-6 expression to determine the concentration of LPS. ELISA and Western blot (WB) were used to detect the PGE2 and CFHR2 expression in each group, respectively. The lentiviral vector for interference and overexpression of the CFHR2 gene was constructed, packaged, and transfected into LPS-induced macrophages. The transfection efficiency was verified by WB. Then, ELISA was used to detect the TNF-α, PGE2, and IL-6 expression. WB was used to detect the CFHR2, iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression. RESULTS The primary isolated cells were identified as macrophages. The LPS-treated macrophages exhibited significantly higher expression of PGE2 and CFHR2, and the inflammatory factors TNF-α and IL-6, as well as iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression compared with the control group (P < 0.05). The TNF-α, PGE2, and IL-6 levels, as well as CFHR2, iNOS, COX-2, TLR2, TLR4, IFN-γ, STAT1, and p-STAT1 expression were considerably lower in the LPS-induced+10% medicated-serum group, LPS-induced+20% medicated-serum group, and shCFHR interference group compared with the LPS group (P < 0.05). CONCLUSION Radix Sophorae Flavescentis might mediate CFHR2 expression and play an important role in inhibiting the LPS-induced pro-inflammatory response of macrophages. Radix Sophorae Flavescentis could be a potential treatment for LPS-induced related inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoying Wu
- Mongolian Medical College, Inner Mongolia Minzu University, Tongliao City, 028000, Inner Mongolia, PR China; Department of Mongolian Medicine, Liaoning Province Mongolian Medicine Hospital, Fuxin City, 123199, Liaoning, PR China.
| | - Li Li
- Second Department of Encephalopathy, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao City, 028007, Inner Mongolia, PR China.
| | - Jinhabure
- Medicated Bath Department, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao City, 028007, Inner Mongolia, PR China.
| | - Xiaofeng
- First Department of Encephalopathy, Affiliated Hospital of Inner Mongolia Minzu University, Tongliao City, 028007, Inner Mongolia, PR China.
| | - Eerdunchaolu
- Mongolian Medical College, Inner Mongolia Minzu University, Tongliao City, 028000, Inner Mongolia, PR China.
| |
Collapse
|
5
|
De Bartolo A, Pasqua T, Romeo N, Rago V, Perrotta I, Giordano F, Granieri MC, Marrone A, Mazza R, Cerra MC, Lefranc B, Leprince J, Anouar Y, Angelone T, Rocca C. The redox-active defensive Selenoprotein T as a novel stress sensor protein playing a key role in the pathophysiology of heart failure. J Transl Med 2024; 22:375. [PMID: 38643121 PMCID: PMC11032602 DOI: 10.1186/s12967-024-05192-w] [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: 02/27/2024] [Accepted: 04/12/2024] [Indexed: 04/22/2024] Open
Abstract
Maladaptive cardiac hypertrophy contributes to the development of heart failure (HF). The oxidoreductase Selenoprotein T (SELENOT) emerged as a key regulator during rat cardiogenesis and acute cardiac protection. However, its action in chronic settings of cardiac dysfunction is not understood. Here, we investigated the role of SELENOT in the pathophysiology of HF: (i) by designing a small peptide (PSELT), recapitulating SELENOT activity via the redox site, and assessed its beneficial action in a preclinical model of HF [aged spontaneously hypertensive heart failure (SHHF) rats] and against isoproterenol (ISO)-induced hypertrophy in rat ventricular H9c2 and adult human AC16 cardiomyocytes; (ii) by evaluating the SELENOT intra-cardiomyocyte production and secretion under hypertrophied stimulation. Results showed that PSELT attenuated systemic inflammation, lipopolysaccharide (LPS)-induced macrophage M1 polarization, myocardial injury, and the severe ultrastructural alterations, while counteracting key mediators of cardiac fibrosis, aging, and DNA damage and restoring desmin downregulation and SELENOT upregulation in the failing hearts. In the hemodynamic assessment, PSELT improved the contractile impairment at baseline and following ischemia/reperfusion injury, and reduced infarct size in normal and failing hearts. At cellular level, PSELT counteracted ISO-mediated hypertrophy and ultrastructural alterations through its redox motif, while mitigating ISO-triggered SELENOT intracellular production and secretion, a phenomenon that presumably reflects the extent of cell damage. Altogether, these results indicate that SELENOT could represent a novel sensor of hypertrophied cardiomyocytes and a potential PSELT-based new therapeutic approach in myocardial hypertrophy and HF.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Ida Perrotta
- Centre for Microscopy and Microanalysis (CM2), Department of Biology, E. and E. S. (DiBEST), University of Calabria, 87036, Rende, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Maria Concetta Granieri
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Alessandro Marrone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Rosa Mazza
- Organ and System Physiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Maria Carmela Cerra
- Organ and System Physiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Benjamin Lefranc
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183, Rouen, France
| | - Jérôme Leprince
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183, Rouen, France
| | - Youssef Anouar
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000, Mont-Saint-Aignan, France
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), 40126, Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
- National Institute of Cardiovascular Research (INRC), 40126, Bologna, Italy
| |
Collapse
|
6
|
Zhang L, Xiu L, Wang T, Zhao D. Effect of L-carnitine in Ameliorating Lipopolysaccharide-Induced Cardiomyocyte Injury via MAPK Signaling. Mol Biotechnol 2024; 66:79-89. [PMID: 37029860 DOI: 10.1007/s12033-023-00731-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023]
Abstract
The present study aimed to elucidate whether L-carnitine (LC) protected H9c2 cells and its underlying mechanisms. Cell counting kit-8 (CCK-8) assay was used to evaluate cell viability. Apoptosis, cell morphology, and lactate dehydrogenase (LDH) assessment were used to prove effects of lipopolysaccharide (LPS) and LC on H9c2 cells. RT-qPCR and western blot assays were hired to evaluate the mRNA and protein expression levels, respectively. ELISA assay was performed to determine the released protein levels. Reactive oxygen species (ROS) level was evaluated by immunofluorescence and flow cytometry. LC was revealed to protect H9c2 cells against LPS-induced injury as indicated by increased cell viability, reduced apoptosis ratio and LDH level. LC treatment also reduced BAX expression as well as up-regulated Bcl-2 expression under LPS treatment. Mechanically, LC reduced oxidative stress and ameliorated the mitochondrial injury through modulating extracellular signal-regulated kinase 1/2 and c-Jun N-terminal protein kinase c-Jun N-terminal protein kinase phosphorylation levels as indicated by decreased membrane potential, increased ATP production and mtDNA expression. We found that LC ameliorates LPS-induced cardiomyocyte injury by abrogating cell apoptosis ratio, ROS levels, as well as mitochondrial dysfunction via mitogen-activated protein kinase signaling. Our findings revealed a potential drug for sepsis or LPS-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- Li Zhang
- Medical College, Internal Medicine Teaching and Research Office, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
- Internal Medicine-Cardiovascular Department, Xinzheng Huaxin Minsheng Hospital, Zhengzhou, Henan, China
| | - Lei Xiu
- Medical College, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
| | - Taoli Wang
- Medical College, Zhengzhou University of Industry Technology, Zhengzhou, Henan, China
| | - Duo Zhao
- Radiology Department, Public People's Hospital of Xinzheng, 2000 Meters South of the Intersection of South China Road and Yanhuang Avenue, Xinzheng, 451100, Henan, China.
| |
Collapse
|
7
|
Hwang YS, Lim J, Yoon HR, Park SH, Kim A, Jang JP, Cho HJ, Lee HG. Astragalus Complanatus Ethanol Attenuates Septic Shock by Exerting Anti-Inflammatory Effects on Macrophages. Int J Mol Sci 2023; 25:384. [PMID: 38203555 PMCID: PMC10778658 DOI: 10.3390/ijms25010384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Sepsis is a systemic inflammatory syndrome that results in multiple-organ failure caused by a dysregulated host immune response to microbial infection. Astragali complanati semen extract (ACSE) exhibits pharmacological activities, including antioxidant, anticancer, antiaging, and anti-diabetes effects. It is widely used in traditional medicine to treat liver and kidney diseases; however, the protective effect of ACSE on sepsis and its mechanisms are unknown. In the present study, we investigated the anti-inflammatory effects and potential mechanisms of the action of ACSE on sepsis. We show that ACSE improved survival rates in mouse models of acute sepsis induced by CLP (cecal ligation and puncture) and LPS stimulation. ACSE administration decreased aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in sepsis-induced mice. Furthermore, ACSE reduced the levels of nitric oxide (NO), tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the serum of septic mice. ACSE treatment inhibited the expression of these proinflammatory genes in LPS-stimulated J774 macrophages. Moreover, ACSE inhibited the phosphorylation of the IκB kinase (IKK) and the nuclear translocation of p65 NF-κB by LPS stimulation in macrophages. These results reveal the mechanism underlying the protective effect of ACSE against sepsis by inhibiting NF-κB activation and suggest that ACSE could be a potential therapeutic candidate to treat acute inflammatory diseases.
Collapse
Affiliation(s)
- Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Jeewon Lim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hyang Ran Yoon
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea;
| | - Aeyung Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea;
| | - Jun-Pil Jang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea;
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon 34141, Republic of Korea; (Y.S.H.); (J.L.); (H.R.Y.)
- Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
8
|
Sun L, Chen Z, Ni Y, He Z. Network pharmacology-based approach to explore the underlying mechanism of sinomenine on sepsis-induced myocardial injury in rats. Front Pharmacol 2023; 14:1138858. [PMID: 37388447 PMCID: PMC10303801 DOI: 10.3389/fphar.2023.1138858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023] Open
Abstract
Background: Sepsis, a systemic disease, usually induces myocardial injury (MI), and sepsis-induced MI has become a significant contributor to sepsis-related deaths in the intensive care unit. The objective of this study is to investigate the role of sinomenine (SIN) on sepsis-induced MI and clarify the underlying mechanism based on the techniques of network pharmacology. Methods: Cecum ligation and puncture (CLP) was adopted to induce sepsis in male Sprague-Dawley (SD) rats. Serum indicators, echocardiographic cardiac parameters, and hematoxylin and eosin (H&E) staining were conducted to gauge the severity of cardiac damage. The candidate targets and potential mechanism of SIN against sepsis-induced MI were analyzed via network pharmacology. Enzyme-linked immunosorbent assay was performed for detecting the serum concentration of inflammatory cytokines. Western blot was applied for evaluating the levels of protein expression. Terminal deoxynucleotidyl transferase-mediated dUTP biotin nick end labeling assay was applied to assess cardiomyocyte apoptosis. Results: SIN significantly improved the cardiac functions, and attenuated myocardial structural damage of rats as compared with the CLP group. In total, 178 targets of SIN and 945 sepsis-related genes were identified, and 33 overlapped targets were considered as candidate targets of SIN against sepsis. Enrichment analysis results demonstrated that these putative targets were significantly associated with the Interleukin 17 (IL-17) signal pathway, inflammatory response, cytokines-mediated signal pathway, and Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) pathway. Molecular docking suggested that SIN had favorable binding affinities with Mitogen-Activated Protein Kinase 8 (MAPK8), Janus Kinase 1 (JAK1), Janus Kinase 2 (JAK2), Signal Transducer and Activator of Transcription 3 (STAT3), and nuclear factor kappa-B (NF-κB). SIN significantly reduced the serum concentration of Tumor Necrosis Factor-α (TNF-α), Interleukin 1 Beta (IL-1β), Interleukin 6 (IL-6), Interferon gamma (IFN-γ), and C-X-C Motif Chemokine Ligand 8 (CXCL8), lowered the protein expression of phosphorylated c-Jun N-terminal kinase 1 (JNK1), JAK1, JAK2, STAT3, NF-κB, and decreased the proportion of cleaved-caspase3/caspase3. In addition, SIN also significantly inhibited the apoptosis of cardiomyocytes as compared with the CLP group. Conclusion: Based on network pharmacology analysis and corresponding experiments, it was concluded that SIN could mediate related targets and pathways to protect against sepsis-induced MI.
Collapse
|
9
|
Peng H, Zhang J, Zhang Z, Turdi S, Han X, Liu Q, Hu H, Ye H, Dong M, Duan Y, Yang Y, Ashrafizadeh M, Rabiee N, Ren J. Cardiac-specific overexpression of catalase attenuates lipopolysaccharide-induced cardiac anomalies through reconciliation of autophagy and ferroptosis. Life Sci 2023:121821. [PMID: 37257582 DOI: 10.1016/j.lfs.2023.121821] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
Lipopolysaccharide (LPS) from Gram-negative bacteria is a major contributor to cardiovascular failure, but the signaling mechanisms underlying its stress response are not fully understood. This study aimed to investigate the effect of the antioxidant enzyme catalase on LPS-induced cardiac abnormalities and the mechanisms involved, with particular focus on the interplay between autophagy, ferroptosis, and apoptosis. Cardiac-specific catalase (CAT) overexpression and wild-type (WT) mice were stimulated with LPS (6 mg/kg, intravenous injection), and cardiac morphology and function were evaluated. Oxidative stress, ferroptosis, apoptosis, and mitochondrial status were monitored, and survival curves were plotted based on the results of LPS stimulation. The results showed that, compared with WT mice, mice overexpressing catalase had a higher survival rate under LPS stimulation. Ultrasound echocardiography, cardiomyocyte characteristics, and Masson's trichrome staining showed that LPS inhibited cardiac function and caused cardiac fibrosis, while catalase alleviated these adverse effects. LPS increased apoptosis (TUNEL, caspase-3 activation, cleaved caspase-3), increased O2·- production, induced inflammation (TNF-α), autophagy, iron toxicity, and carbonyl damage, and significantly damaged mitochondria (mitochondrial membrane potential, mitochondrial proteins, and ultrastructure). These effects were significantly alleviated by catalase. Interestingly, the antioxidant N-acetylcysteine, autophagy inhibitor 3-methyladenine, and ferroptosis inhibitor lipostatin-1 all eliminated the LPS-induced contraction dysfunction and ferroptosis (using lipid peroxidation). Induction of ferroptosis could eliminate the cardioprotective effect of NAC. In conclusion, catalase rescues LPS-induced cardiac dysfunction by regulating oxidative stress, autophagy, ferroptosis, apoptosis, and mitochondrial damage in cardiomyocytes.
Collapse
Affiliation(s)
- Hu Peng
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China.
| | - Ji Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Zhonglin Zhang
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Subat Turdi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Xuefeng Han
- Department of Physiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi 710069, China
| | - Huantao Hu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hua Ye
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, China
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yu Duan
- Department of Cardiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi 710069, China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, School of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi 710069, China
| | - Milad Ashrafizadeh
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China; Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Navid Rabiee
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai 200032, China.
| |
Collapse
|
10
|
Nong Y, Wei X, Yu D. Inflammatory mechanisms and intervention strategies for sepsis-induced myocardial dysfunction. Immun Inflamm Dis 2023; 11:e860. [PMID: 37249297 PMCID: PMC10187025 DOI: 10.1002/iid3.860] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/30/2022] [Accepted: 04/21/2023] [Indexed: 05/31/2023] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is the leading cause of death in patients with sepsis in the intensive care units. The main manifestations of SIMD are systolic and diastolic dysfunctions of the myocardium. Despite our initial understanding of the SIMD over the past three decades, the incidence and mortality of SIMD remain high. This may be attributed to the large degree of heterogeneity among the initiating factors, disease processes, and host states involved in SIMD. Previously, organ dysfunction caused by sepsis was thought to be an impairment brought about by an excessive inflammatory response. However, many recent studies have shown that SIMD is a consequence of a combination of factors shaped by the inflammatory responses between the pathogen and the host. In this article, we review the mechanisms of the inflammatory responses and potential novel therapeutic strategies in SIMD.
Collapse
Affiliation(s)
- Yuxin Nong
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xuebiao Wei
- Department of Geriatric Intensive Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Danqing Yu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| |
Collapse
|
11
|
Rocca C, De Bartolo A, Guzzi R, Crocco MC, Rago V, Romeo N, Perrotta I, De Francesco EM, Muoio MG, Granieri MC, Pasqua T, Mazza R, Boukhzar L, Lefranc B, Leprince J, Gallo Cantafio ME, Soda T, Amodio N, Anouar Y, Angelone T. Palmitate-Induced Cardiac Lipotoxicity Is Relieved by the Redox-Active Motif of SELENOT through Improving Mitochondrial Function and Regulating Metabolic State. Cells 2023; 12:cells12071042. [PMID: 37048116 PMCID: PMC10093731 DOI: 10.3390/cells12071042] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Cardiac lipotoxicity is an important contributor to cardiovascular complications during obesity. Given the fundamental role of the endoplasmic reticulum (ER)-resident Selenoprotein T (SELENOT) for cardiomyocyte differentiation and protection and for the regulation of glucose metabolism, we took advantage of a small peptide (PSELT), derived from the SELENOT redox-active motif, to uncover the mechanisms through which PSELT could protect cardiomyocytes against lipotoxicity. To this aim, we modeled cardiac lipotoxicity by exposing H9c2 cardiomyocytes to palmitate (PA). The results showed that PSELT counteracted PA-induced cell death, lactate dehydrogenase release, and the accumulation of intracellular lipid droplets, while an inert form of the peptide (I-PSELT) lacking selenocysteine was not active against PA-induced cardiomyocyte death. Mechanistically, PSELT counteracted PA-induced cytosolic and mitochondrial oxidative stress and rescued SELENOT expression that was downregulated by PA through FAT/CD36 (cluster of differentiation 36/fatty acid translocase), the main transporter of fatty acids in the heart. Immunofluorescence analysis indicated that PSELT also relieved the PA-dependent increase in CD36 expression, while in SELENOT-deficient cardiomyocytes, PA exacerbated cell death, which was not mitigated by exogenous PSELT. On the other hand, PSELT improved mitochondrial respiration during PA treatment and regulated mitochondrial biogenesis and dynamics, preventing the PA-provoked decrease in PGC1-α and increase in DRP-1 and OPA-1. These findings were corroborated by transmission electron microscopy (TEM), revealing that PSELT improved the cardiomyocyte and mitochondrial ultrastructures and restored the ER network. Spectroscopic characterization indicated that PSELT significantly attenuated infrared spectral-related macromolecular changes (i.e., content of lipids, proteins, nucleic acids, and carbohydrates) and also prevented the decrease in membrane fluidity induced by PA. Our findings further delineate the biological significance of SELENOT in cardiomyocytes and indicate the potential of its mimetic PSELT as a protective agent for counteracting cardiac lipotoxicity.
Collapse
Affiliation(s)
- Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Anna De Bartolo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
| | - Rita Guzzi
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036 Rende, Italy
- CNR-NANOTEC, Department of Physics, University of Calabria, 87036 Rende, Italy
| | - Maria Caterina Crocco
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036 Rende, Italy
- STAR Research Infrastructure, University of Calabria, Via Tito Flavio, 87036 Rende, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Naomi Romeo
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Ida Perrotta
- Centre for Microscopy and Microanalysis (CM2), Department of Biology, Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95124 Catania, Italy
| | - Maria Grazia Muoio
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95124 Catania, Italy
| | - Maria Concetta Granieri
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Teresa Pasqua
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Rosa Mazza
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Loubna Boukhzar
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
| | - Benjamin Lefranc
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | - Jérôme Leprince
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | | | - Teresa Soda
- Department of Health Science, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Youssef Anouar
- UNIROUEN, Inserm U1239, Neuroendocrine, Endocrine and Germinal Differentiation and Communication (NorDiC), Rouen Normandie University, 76000 Mont-Saint-Aignan, France
- UNIROUEN, UMS-UAR HERACLES, PRIMACEN, Cell Imaging Platform of Normandy, Institute for Research and Innovation in Biomedicine (IRIB), 76183 Rouen, France
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, 87036 Rende, Italy
- National Institute of Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
12
|
Granieri MC, Rocca C, De Bartolo A, Nettore IC, Rago V, Romeo N, Ceramella J, Mariconda A, Macchia PE, Ungaro P, Sinicropi MS, Angelone T. Quercetin and Its Derivative Counteract Palmitate-Dependent Lipotoxicity by Inhibiting Oxidative Stress and Inflammation in Cardiomyocytes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3492. [PMID: 36834186 PMCID: PMC9958705 DOI: 10.3390/ijerph20043492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 06/18/2023]
Abstract
Cardiac lipotoxicity plays an important role in the pathogenesis of obesity-related cardiovascular disease. The flavonoid quercetin (QUE), a nutraceutical compound that is abundant in the "Mediterranean diet", has been shown to be a potential therapeutic agent in cardiac and metabolic diseases. Here, we investigated the beneficial role of QUE and its derivative Q2, which demonstrates improved bioavailability and chemical stability, in cardiac lipotoxicity. To this end, H9c2 cardiomyocytes were pre-treated with QUE or Q2 and then exposed to palmitate (PA) to recapitulate the cardiac lipotoxicity occurring in obesity. Our results showed that both QUE and Q2 significantly attenuated PA-dependent cell death, although QUE was effective at a lower concentration (50 nM) when compared with Q2 (250 nM). QUE decreased the release of lactate dehydrogenase (LDH), an important indicator of cytotoxicity, and the accumulation of intracellular lipid droplets triggered by PA. On the other hand, QUE protected cardiomyocytes from PA-induced oxidative stress by counteracting the formation of malondialdehyde (MDA) and protein carbonyl groups (which are indicators of lipid peroxidation and protein oxidation, respectively) and intracellular ROS generation, and by improving the enzymatic activities of catalase and superoxide dismutase (SOD). Pre-treatment with QUE also significantly attenuated the inflammatory response induced by PA by reducing the release of key proinflammatory cytokines (IL-1β and TNF-α). Similar to QUE, Q2 (250 nM) also significantly counteracted the PA-provoked increase in intracellular lipid droplets, LDH, and MDA, improving SOD activity and decreasing the release of IL-1β and TNF-α. These results suggest that QUE and Q2 could be considered potential therapeutics for the treatment of the cardiac lipotoxicity that occurs in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Maria Concetta Granieri
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Naomi Romeo
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Annaluisa Mariconda
- Department of Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Scuola di Medicina, Università degli Studi di Napoli Federico II, 80131 Naples, Italy
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale (IEOS) “Gaetano Salvatore”, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Pathophysiology, Department of Biology, Ecology and Earth Science (DiBEST), University of Calabria, 87036 Rende, Italy
- National Institute of Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
13
|
Jiang T, Wang Q, Lv J, Lin L. Mitochondria-endoplasmic reticulum contacts in sepsis-induced myocardial dysfunction. Front Cell Dev Biol 2022; 10:1036225. [PMID: 36506093 PMCID: PMC9730255 DOI: 10.3389/fcell.2022.1036225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Mitochondrial and endoplasmic reticulum (ER) are important intracellular organelles. The sites that mitochondrial and ER are closely related in structure and function are called Mitochondria-ER contacts (MERCs). MERCs are involved in a variety of biological processes, including calcium signaling, lipid synthesis and transport, autophagy, mitochondrial dynamics, ER stress, and inflammation. Sepsis-induced myocardial dysfunction (SIMD) is a vital organ damage caused by sepsis, which is closely associated with mitochondrial and ER dysfunction. Growing evidence strongly supports the role of MERCs in the pathogenesis of SIMD. In this review, we summarize the biological functions of MERCs and the roles of MERCs proteins in SIMD.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jiagao Lv, ; Li Lin, ,
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Jiagao Lv, ; Li Lin, ,
| |
Collapse
|
14
|
Omaveloxolone attenuates the sepsis-induced cardiomyopathy via activating the nuclear factor erythroid 2-related factor 2. Int Immunopharmacol 2022; 111:109067. [PMID: 35908503 DOI: 10.1016/j.intimp.2022.109067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022]
Abstract
Sepsis-induced cardiomyopathy (SIC) is a common complication of sepsis and is the main reason for the high mortality in sepsis patients. More recent studies have indicated that activating nuclear factor erythroid 2-related factor 2 (Nrf2) signaling plays a protective role in SIC. As a potent activator of Nrf2, Omaveloxolone plays a pivotal role in defending against oxidative stress and the inflammatory response. Thus, we examined the efficacy of omaveloxolone in SIC. In the present study, the mice were injected intraperitoneally with a single dose of LPS (10 mg/kg) for 12 h to induce SIC. The data in our study indicated that omaveloxolone administration significantly improved cardiac injury and dysfunction in LPS-induced SIC. In addition, omaveloxolone administration reduced SIC-related cardiac oxidative stress, the inflammatory response and cardiomyocyte apoptosis in mice. In addition, omaveloxolone administration also improved LPS-induced cardiomyocyte injury in an in vitro model using H9C2 cells. Moreover, knockdown of Nrf2 by si-Nrf2 abolished the omaveloxolone-mediated cardioprotective effects. In conclusion, omaveloxolone has potent cardioprotective potential in treating sepsis and SIC via activation of the Nrf2 signaling pathway.
Collapse
|
15
|
Peng Y, Wang L, Zhao X, Lai S, He X, Fan Q, He H, He M. Puerarin attenuates lipopolysaccharide-induced myocardial injury via the 14-3-3γ/PKCε pathway activating adaptive autophagy. Int Immunopharmacol 2022; 108:108905. [DOI: 10.1016/j.intimp.2022.108905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 12/30/2022]
|
16
|
The Antioxidant Selenoprotein T Mimetic, PSELT, Induces Preconditioning-like Myocardial Protection by Relieving Endoplasmic-Reticulum Stress. Antioxidants (Basel) 2022; 11:antiox11030571. [PMID: 35326221 PMCID: PMC8944960 DOI: 10.3390/antiox11030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress and endoplasmic reticulum stress (ERS) are strictly involved in myocardial ischemia/reperfusion (MI/R). Selenoprotein T (SELENOT), a vital thioredoxin-like selenoprotein, is crucial for ER homeostasis and cardiomyocyte differentiation and protection, likely acting as a redox-sensing protein during MI/R. Here, we designed a small peptide (PSELT), encompassing the redox site of SELENOT, and investigated whether its pre-conditioning cardioprotective effect resulted from modulating ERS during I/R. The Langendorff rat heart model was employed for hemodynamic analysis, while mechanistic studies were performed in perfused hearts and H9c2 cardiomyoblasts. PSELT improved the post-ischemic contractile recovery, reducing infarct size and LDH release with and without the ERS inducer tunicamycin (TM). Mechanistically, I/R and TM upregulated SELENOT expression, which was further enhanced by PSELT. PSELT also prevented the expression of the ERS markers CHOP and ATF6, reduced cardiac lipid peroxidation and protein oxidation, and increased SOD and catalase activities. An inert PSELT (I-PSELT) lacking selenocysteine was ineffective. In H9c2 cells, H2O2 decreased cell viability and SELENOT expression, while PSELT rescued protein levels protecting against cell death. In SELENOT-deficient H9c2 cells, H2O2 exacerbated cell death, that was partially mitigated by PSELT. Microscopy analysis revealed that a fluorescent form of PSELT was internalized into cardiomyocytes with a perinuclear distribution. Conclusions: The cell-permeable PSELT is able to induce pharmacological preconditioning cardioprotection by mitigating ERS and oxidative stress, and by regulating endogenous SELENOT.
Collapse
|
17
|
Mancino D, Kharouf N, Scavello F, Hellé S, Salloum-Yared F, Mutschler A, Mathieu E, Lavalle P, Metz-Boutigue MH, Haïkel Y. The Catestatin-Derived Peptides Are New Actors to Fight the Development of Oral Candidosis. Int J Mol Sci 2022; 23:ijms23042066. [PMID: 35216181 PMCID: PMC8876135 DOI: 10.3390/ijms23042066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
Resistance to antifungal therapy of Candida albicans and non-albicans Candida strains, frequently associated with oral candidosis, is on the rise. In this context, host-defense peptides have emerged as new promising candidates to overcome antifungal resistance. Thus, the aim of this study was to assess the effectiveness against Candida species of different Catestatin-derived peptides, as well as the combined effect with serum albumin. Among Catestatin-derived peptides, the most active against sensitive and resistant strains of C. albicans, C. tropicalis and C. glabrata was the D-isomer of Cateslytin (D-bCtl) whereas the efficiency of the L-isomer (L-bCtl) significantly decreases against C. glabrata strains. Images obtained by transmission electron microscopy clearly demonstrated fungal membrane lysis and the leakage of the intracellular material induced by the L-bCtl and D-bCtl peptides. The possible synergistic effect of albumin on Catestatin-derived peptides activity was investigated too. Our finding showed that bovine serum albumin (BSA) when combined with the L- isomer of Catestatin (L-bCts) had a synergistic effect against Candida albicans especially at low concentrations of BSA; however, no synergistic effect was detected when BSA interacted with L-bCtl, suggesting the importance of the C-terminal end of L-bCts (GPGLQL) for the interaction with BSA. In this context in vitro D-bCtl, as well as the combination of BSA with L-bCts are potential candidates for the development of new antifungal drugs for the treatment of oral candidosis due to Candida and non-Candida albicans, without detrimental side effects.
Collapse
Affiliation(s)
- Davide Mancino
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, University of Strasbourg, 67000 Strasbourg, France
| | - Naji Kharouf
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Correspondence: ; Tel.: +33-66752-2841
| | - Francesco Scavello
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Sophie Hellé
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Fouad Salloum-Yared
- Department of Medical Laboratory, The General Authority of the Syrian Arab Red Crescent Hospital, Damascus 0100, Syria;
| | - Angela Mutschler
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Eric Mathieu
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Philippe Lavalle
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Marie-Hélène Metz-Boutigue
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
| | - Youssef Haïkel
- Department of Biomaterials and Bioengineering, INSERM UMR_S 1121, University of Strasbourg, 67000 Strasbourg, France; (D.M.); (F.S.); (S.H.); (A.M.); (E.M.); (P.L.); (M.-H.M.-B.); (Y.H.)
- Department of Endodontics and Conservative Dentistry, Faculty of Dental Medicine, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
18
|
Scavello F, Mutschler A, Hellé S, Schneider F, Chasserot-Golaz S, Strub JM, Cianferani S, Haikel Y, Metz-Boutigue MH. Catestatin in innate immunity and Cateslytin-derived peptides against superbugs. Sci Rep 2021; 11:15615. [PMID: 34341386 PMCID: PMC8329280 DOI: 10.1038/s41598-021-94749-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
Chromogranin A (CgA) is the precursor of several antimicrobial peptides, such as Catestatin (Cts, bovine CgA344-364), initially described as a potent inhibitor of catecholamines. This peptide displays direct antimicrobial activities and contributes to immune system regulation. The aim of the present study is to investigate a designed peptide based on Cts to fight infections against superbugs and more particularly Staphylococcus aureus. In addition to Cateslytin (Ctl, bovine CgA344-358), the active domain of Catestatin, several peptides including dimers, D-isomer and the new designed peptide DOPA-K-DOPA-K-DOPA-TLRGGE-RSMRLSFRARGYGFR (Dopa5T-Ctl) were prepared and tested. Cateslytin is resistant to bacterial degradation and does not induce bacterial resistance. The interaction of Catestatin with immune dermal cells (dendritic cells DC1a, dermal macrophages CD14 and macrophages) was analyzed by using confocal microscopy and cytokine release assay. The dimers and D-isomer of Ctl were tested against a large variety of bacteria showing the potent antibacterial activity of the D-isomer. The peptide Dopa5T-Ctl is able to induce the self-killing of S. aureus after release of Ctl by the endoprotease Glu-C produced by this pathogen. It permits localized on-demand delivery of the antimicrobial drug directly at the infectious site.
Collapse
Affiliation(s)
- Francesco Scavello
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France
- Department of Biology, Ecology and Earth Science, University of Calabria, Arcavacata di Rende, Italy
| | - Angela Mutschler
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France
| | - Sophie Hellé
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France
- Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Francis Schneider
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France
- Faculty of Medicine, University of Strasbourg, Strasbourg, France
- Médecine Intensive-Réanimation, Hautepierre Hospital, Hôpitaux Universitaires, Strasbourg, Federation of Translational Medicine, Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Sylvette Chasserot-Golaz
- Centre National de la Recherche Scientifique, Institut des Neurosciences Cellulaires et Intégratives, University of Strasbourg, Strasbourg, France
| | - Jean-Marc Strub
- Centre National de la Recherche Scientifique, Laboratory of Bio-Organic Mass Spectrometry, Analytical Sciences Department, Pluridisciplinary Institute Hubert Curien, UMR 7178, University of Strasbourg, Strasbourg, France
| | - Sarah Cianferani
- Centre National de la Recherche Scientifique, Laboratory of Bio-Organic Mass Spectrometry, Analytical Sciences Department, Pluridisciplinary Institute Hubert Curien, UMR 7178, University of Strasbourg, Strasbourg, France
| | - Youssef Haikel
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France
- Faculty of Odontology, University of Strasbourg, Strasbourg, France
| | - Marie-Hélène Metz-Boutigue
- BioMaterials and BioEngeneering, Institut National de la Santé et de la Recherche Médicale UMR_S 1121, Federation of Translational Medicine Faculty, of Odontology, University of Strasbourg, Hôpital Civil, Porte de L'Hôpital, 67000, Strasbourg, France.
| |
Collapse
|
19
|
The Emerging Roles of Chromogranins and Derived Polypeptides in Atherosclerosis, Diabetes, and Coronary Heart Disease. Int J Mol Sci 2021; 22:ijms22116118. [PMID: 34204153 PMCID: PMC8201018 DOI: 10.3390/ijms22116118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Chromogranin A (CgA), B (CgB), and C (CgC), the family members of the granin glycoproteins, are associated with diabetes. These proteins are abundantly expressed in neurons, endocrine, and neuroendocrine cells. They are also present in other areas of the body. Patients with diabetic retinopathy have higher levels of CgA, CgB, and CgC in the vitreous humor. In addition, type 1 diabetic patients have high CgA and low CgB levels in the circulating blood. Plasma CgA levels are increased in patients with hypertension, coronary heart disease, and heart failure. CgA is the precursor to several functional peptides, including catestatin, vasostatin-1, vasostatin-2, pancreastatin, chromofungin, and many others. Catestatin, vasostain-1, and vasostatin-2 suppress the expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in human vascular endothelial cells. Catestatin and vasostatin-1 suppress oxidized low-density lipoprotein-induced foam cell formation in human macrophages. Catestatin and vasostatin-2, but not vasostatin-1, suppress the proliferation and these three peptides suppress the migration in human vascular smooth muscles. Chronic infusion of catestatin, vasostatin-1, or vasostatin-2 suppresses the development of atherosclerosis of the aorta in apolipoprotein E-deficient mice. Catestatin, vasostatin-1, vasostatin-2, and chromofungin protect ischemia/reperfusion-induced myocardial dysfunction in rats. Since pancreastatin inhibits insulin secretion from pancreatic β-cells, and regulates glucose metabolism in liver and adipose tissues, pancreastatin inhibitor peptide-8 (PSTi8) improves insulin resistance and glucose homeostasis. Catestatin stimulates therapeutic angiogenesis in the mouse hind limb ischemia model. Gene therapy with secretoneurin, a CgC-derived peptide, stimulates postischemic neovascularization in apolipoprotein E-deficient mice and streptozotocin-induced diabetic mice, and improves diabetic neuropathy in db/db mice. Therefore, CgA is a biomarker for atherosclerosis, diabetes, hypertension, and coronary heart disease. CgA- and CgC--derived polypeptides provide the therapeutic target for atherosclerosis and ischemia-induced tissue damages. PSTi8 is useful in the treatment of diabetes.
Collapse
|