1
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Overview of pyroptosis mechanism and in-depth analysis of cardiomyocyte pyroptosis mediated by NF-κB pathway in heart failure. Biomed Pharmacother 2024; 179:117367. [PMID: 39214011 DOI: 10.1016/j.biopha.2024.117367] [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: 07/17/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
The pyroptosis of cardiomyocytes has become an essential topic in heart failure research. The abnormal accumulation of these biological factors, including angiotensin II, advanced glycation end products, and various growth factors (such as connective tissue growth factor, vascular endothelial growth factor, transforming growth factor beta, among others), activates the nuclear factor-κB (NF-κB) signaling pathway in cardiovascular diseases, ultimately leading to pyroptosis of cardiomyocytes. Therefore, exploring the underlying molecular biological mechanisms is essential for developing novel drugs and therapeutic strategies. However, our current understanding of the precise regulatory mechanism of this complex signaling pathway in cardiomyocyte pyroptosis is still limited. Given this, this study reviews the milestone discoveries in the field of pyroptosis research since 1986, analyzes in detail the similarities, differences, and interactions between pyroptosis and other cell death modes (such as apoptosis, necroptosis, autophagy, and ferroptosis), and explores the deep connection between pyroptosis and heart failure. At the same time, it depicts in detail the complete pathway of the activation, transmission, and eventual cardiomyocyte pyroptosis of the NF-κB signaling pathway in the process of heart failure. In addition, the study also systematically summarizes various therapeutic approaches that can inhibit NF-κB to reduce cardiomyocyte pyroptosis, including drugs, natural compounds, small molecule inhibitors, gene editing, and other cutting-edge technologies, aiming to provide solid scientific support and new research perspectives for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
2
|
Özbay Kurt FG, Cicortas BA, Balzasch BM, De la Torre C, Ast V, Tavukcuoglu E, Ak C, Wohlfeil SA, Cerwenka A, Utikal J, Umansky V. S100A9 and HMGB1 orchestrate MDSC-mediated immunosuppression in melanoma through TLR4 signaling. J Immunother Cancer 2024; 12:e009552. [PMID: 39266214 PMCID: PMC11409250 DOI: 10.1136/jitc-2024-009552] [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] [Accepted: 08/01/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Immunotherapies for malignant melanoma are challenged by the resistance developed in a significant proportion of patients. Myeloid-derived suppressor cells (MDSC), with their ability to inhibit antitumor T-cell responses, are a major contributor to immunosuppression and resistance to immune checkpoint therapies in melanoma. Damage-associated molecular patterns S100A8, S100A9, and HMGB1, acting as toll like receptor 4 (TLR4) and receptor for advanced glycation endproducts (RAGE) ligands, are highly expressed in the tumor microenvironment and drive MDSC activation. However, the role of TLR4 and RAGE signaling in the acquisition of MDSC immunosuppressive properties remains to be better defined. Our study investigates how the signaling via TLR4 and RAGE as well as their ligands S100A9 and HMGB1, shape MDSC-mediated immunosuppression in melanoma. METHODS MDSC were isolated from the peripheral blood of patients with advanced melanoma or generated in vitro from healthy donor-derived monocytes. Monocytes were treated with S100A9 or HMGB1 for 72 hours. The immunosuppressive capacity of treated monocytes was assessed in the inhibition of T-cell proliferation assay in the presence or absence of TLR4 and RAGE inhibitors. Plasma levels of S100A8/9 and HMGB1 were quantified by ELISA. Single-cell RNA sequencing (scRNA-seq) was performed on monocytes from patients with melanoma and healthy donors. RESULTS We showed that exposure to S100A9 and HMGB1 converted healthy donor-derived monocytes into MDSC through TLR4 signaling. Our scRNA-seq data revealed in patient monocytes enriched inflammatory genes, including S100 and those involved in NF-κB and TLR4 signaling, and a reduced major histocompatibility complex II gene expression. Furthermore, elevated plasma S100A8/9 levels correlated with shorter progression-free survival in patients with melanoma. CONCLUSIONS These findings highlight the critical role of TLR4 and, to a lesser extent, RAGE signaling in the conversion of monocytes into MDSC-like cells, underscore the potential of targeting S100A9 to prevent this conversion, and highlight the prognostic value of S100A8/9 as a plasma biomarker in melanoma.
Collapse
Affiliation(s)
- Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Beatrice-Ana Cicortas
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bianca M Balzasch
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carolina De la Torre
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Volker Ast
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ece Tavukcuoglu
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Cagla Ak
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sebastian A Wohlfeil
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany
- Department of Dermatology Venereology and Allergology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
3
|
Hassan NF, El-Ansary MR, Selim HMRM, Ousman MS, Khattab MS, El-Ansary MRM, Gad ES, Moursi SMM, Gohar A, Gowifel AMH. Alirocumab boosts antioxidant status and halts inflammation in rat model of sepsis-induced nephrotoxicity via modulation of Nrf2/HO-1, PCSK9/HMGB1/NF-ᴋB/NLRP3 and Fractalkine/CX3CR1 hubs. Biomed Pharmacother 2024; 177:116929. [PMID: 38889644 DOI: 10.1016/j.biopha.2024.116929] [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: 04/17/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
Acute kidney injury (AKI) is a devastating consequence of sepsis, accompanied by high mortality rates. It was suggested that inflammatory pathways are closely linked to the pathogenesis of lipopolysaccharide (LPS)-induced AKI. Inflammatory signaling, including PCSK9, HMGB1/RAGE/TLR4/MYD88/NF-κB, NLRP3/caspase-1 and Fractalkine/CX3CR1 are considered major forerunners in this link. Alirocumab, PCSK9 inhibitor, with remarkable anti-inflammatory features. Accordingly, this study aimed to elucidate the antibacterial effect of alirocumab against E. coli in vitro. Additionally, evaluation of the potential nephroprotective effects of alirocumab against LPS-induced AKI in rats, highlighting the potential underlying mechanisms involved in these beneficial actions. Thirty-six adult male Wistar rats were assorted into three groups (n=12). Group I; was a normal control group, whereas sepsis-mediated AKI was induced in groups II and III through single-dose intraperitoneal injection of LPS on day 16. In group III, animals were given alirocumab. The results revealed that LPS-induced AKI was mitigated by alirocumab, evidenced by amelioration in renal function tests (creatinine, cystatin C, KIM-1, and NGAL); oxidative stress biomarkers (Nrf2, HO-1, TAC, and MDA); apoptotic markers and renal histopathological findings. Besides, alirocumab pronouncedly hindered LPS-mediated inflammatory response, confirmed by diminishing HMGB1, TNF-α, IL-1β, and caspase-1 contents; the gene expression of PCSK9, RAGE, NF-ᴋB and Fractalkine/CX3CR1, along with mRNA expression of TLR4, MYD88, and NLRP3. Regarding the antibacterial actions, results showed that alirocumab displayed potential anti-bacterial activity against pathogenic gram-negative E. coli. In conclusion, alirocumab elicited nephroprotective activities against LPS-induced AKI via modulation of Nrf2/HO-1, PCSK9, HMGB1/RAGE/TLR4/MYD88/NF-ᴋB/NLRP3/Caspase-1, Fractalkine/CX3R1 and apoptotic axes.
Collapse
Affiliation(s)
- Noha F Hassan
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Mona R El-Ansary
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Heba Mohammed Refat M Selim
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University, Cairo 11651, Egypt.
| | - Mona S Ousman
- Emergency Medical Services, College of Applied Sciences, AlMaarefa University, P.O. Box 71666, Riyadh, Saudi Arabia.
| | - Marwa S Khattab
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza 1211, Egypt.
| | - Mahmoud R M El-Ansary
- Medical Microbiology and Immunology Department, Faculty of Medicine, Misr University for Science and Technology (MUST), Giza 12566, Egypt.
| | - Enas S Gad
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Pharmacology and Toxicology, faculty of Pharmacy, Sinai University-Kantara branch, Ismailia, Egypt
| | - Suzan M M Moursi
- Medical Physiology Department, Faculty of Medicine, Zagazig University, 44519, Egypt.
| | - Asmaa Gohar
- Microbiology and Immunology Department, Faculty of Pharmacy, Ahram Canadian University, sixth of October city, Giza, Egypt; Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala City, Suez, 43713, Egypt.
| | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| |
Collapse
|
4
|
Tian H, Chen H, Yin X, Lv M, Wei L, Zhang Y, Jia S, Li J, Song H. CORM-3 Inhibits the Inflammatory Response of Human Periodontal Ligament Fibroblasts Stimulated by LPS and High Glucose. J Inflamm Res 2024; 17:4845-4863. [PMID: 39070135 PMCID: PMC11277920 DOI: 10.2147/jir.s460954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/18/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Diabetes has been recognized as an independent risk factor for periodontitis. Increasing evidences indicate that hyperglycemia aggravates inflammatory response of human periodontal ligament cells (hPDLCs). Carbon monoxide-releasing molecule-3 (CORM-3) is a water-soluble compound that can release carbon monoxide (CO) in a controllable manner. CORM-3 has been shown the anti-inflammatory effect in different cell lineages. Methods We stimulated periodontal ligament cells with LPS and high glucose. The expression of inflammatory cytokine was detected by ELISA. RT-qPCR, Western blot and immunofluorescence were used to detect the expression of TLR2, TLR4, RAGE and the activation of NF-κB pathway. We performed silencing and overexpression treatment of RAGE targeting the role of RAGE. We performed the immunostaining of paraffin sections of the periodontitis model in diabetes rats. Results The results showed that CORM-3 significantly inhibited the expression of inflammatory cytokine in hPDLCs stimulated with LPS and high glucose. CORM-3 also inhibited LPS and high glucose-induced expression of RAGE/NF-κB pathway and TLR2/TLR4/NF-κB pathway. Silence of RAGE resulted in significantly decreased expression of proteins above. Overexpression of RAGE significantly enhanced the expression of these factors. CORM-3 abrogated the effect of RAGE partially. In animal model, CORM-3 suppressed the inflammatory response of periodontal tissues in experimental periodontitis of diabetic rats. Discussion Our research proved CORM-3 reduced the inflammatory response via RAGE/NF-κB pathway and TLR2/TLR4/NF-κB pathway in the process of high glucose exacerbated periodontitis. These findings demonstrated the role of RAGE in the process of high glucose exacerbated periodontitis and suggested that CORM3 be a potential therapeutic strategy for the treatment of diabetes patients with periodontitis.
Collapse
Affiliation(s)
- Haoyang Tian
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, People’s Republic of China
| | - Hui Chen
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, People’s Republic of China
| | - Xiaochun Yin
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, People’s Republic of China
| | - Meiyi Lv
- Department of Pediatric Dentistry, Jinan Stomatological Hospital, Jinan, People’s Republic of China
| | - Lingling Wei
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, People’s Republic of China
| | - Yuna Zhang
- Department of Stomatology, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Shuhan Jia
- Department of Stomatology, Yancheng NO. 1 People’s Hospital, Yancheng, People’s Republic of China
| | - Jingyuan Li
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, People’s Republic of China
| | - Hui Song
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, People’s Republic of China
| |
Collapse
|
5
|
Zodio S, Serreli G, Melis MP, Franchi B, Boronat A, de la Torre R, Deiana M. Protective effect of hydroxytyrosol and tyrosol metabolites in LPS-induced vascular barrier derangement in vitro. Front Nutr 2024; 11:1350378. [PMID: 38706564 PMCID: PMC11066181 DOI: 10.3389/fnut.2024.1350378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction The maintenance of endothelial barrier function is essential for vasal homeostasis and prevention of cardiovascular diseases. Among the toxic stimuli involved in the initiation of atherosclerotic lesions, Gram negative lipopolysaccharide (LPS) has been reported to be able to trigger endothelial dysfunction, through the alteration of barrier permeability and inflammatory response. Hydroxytyrosol (HT) and tyrosol (Tyr), the major phenolic compounds of extra virgin olive oil (EVOO), as wells as their circulating sulphated and glucuronidated metabolites have been shown to exert anti-inflammatory effects at endothelial level. Methods In this study we investigated the protective effects of HT and Tyr metabolites on LPS-induced alteration of permeability in Human Umbilical Vein Endothelial Cells (HUVEC) monolayers and examined underlying signaling pathways, focusing on tight junction (TJ) proteins, mitogen-activated protein kinase (MAPK) and NOD-, LRR-and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. Results It was shown that LPS-increased permeability in HUVEC cells was due to the alteration of TJ protein level, following the activation of MAPK and NLRP3. HT and Tyr sulphated and glucuronidated metabolites were able to limit the effects exerted by LPS, acting as signaling molecules with an efficacy comparable to that of their precursors HT and Tyr. Discussion The obtained results add a further piece to the understanding of HT and Tyr metabolites mechanisms of action in vascular protection.
Collapse
Affiliation(s)
- Sonia Zodio
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Gabriele Serreli
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Maria Paola Melis
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Benedetta Franchi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Anna Boronat
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Integrative Pharmacology and Systems Neurosciences Research Group, Hospital del Mar Research Institute, Barcelona, Spain
| | - Rafael de la Torre
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Integrative Pharmacology and Systems Neurosciences Research Group, Hospital del Mar Research Institute, Barcelona, Spain
- Physiopathology of Obesity and Nutrition Networking Biomedical Research Centre (CIBEROBN), Madrid, Spain
| | - Monica Deiana
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
6
|
Apte MM, Khattar E, Tupe RS. Mechanistic role of Syzygium cumini (L.) Skeels in glycation induced diabetic nephropathy via RAGE-NF-κB pathway and extracellular proteins modifications: A molecular approach. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117573. [PMID: 38110133 DOI: 10.1016/j.jep.2023.117573] [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: 10/02/2023] [Revised: 11/23/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Syzygium cumini (L.) Skeels (SC), an ancient medicinal plant, is used as a complementary and alternative medicine for treating diabetes mellitus and its associated complications, such as diabetic nephropathy (DN). Phytochemicals present in SC homeopathic formulations possess anti-glycemic, anti-glycation, anti-inflammatory, and antioxidant properties. Additionally, the non-enzymatic formation of advanced glycation end products (AGEs) increases during hyperglycemia in diabetes. AGEs interaction with their receptor of AGEs (RAGE) promotes inflammation via Nuclear Factor-κB (NF-κB) and the accumulation of Extracellular Matrix (ECM) proteins, contributing to the renal dysfunction in DN. However, the molecular mechanism through which SC formulations interact with the AGEs-RAGE-NF-κB pathway has not yet been investigated. AIM This study aims to examine the impact of SC formulations on the RAGE-NF-κB pathway and ECM protein modifications in glycation-induced DN using a molecular approach. MATERIALS AND METHODS Human serum albumin (10 mg/ml) was glycated with MGO (55 mM) in the presence of SC formulations - Mother tincture (MT), 30C, 200C for 7 days. Glycated samples were added to renal cells (HEK 293) for 24 h. Subsequently, cellular gene and protein expressions of RAGE, NF-κB, vascular endothelial growth factor (VEGF), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), collagen IV (Col IV), and fibronectin were determined using RT-qPCR and Western blot analysis. The immunofluorescence, luciferase assay, and chromatin immunoprecipitation techniques were employed to gain insights into glycation-induced NF-κB nuclear translocation, transcriptional activity, and its effect on RAGE promoter activity in SC-treated cells. RESULTS SC formulations significantly downregulated glycation-induced elevated levels of RAGE and NF-κB. Mechanistically, SC formulations prevented NF-κB nuclear translocation, transcriptional activity, and RAGE promoter activity. Also, SC formulations significantly attenuated glycation-enhanced expressions of inflammatory cytokines (IL-6, TNF-α, and VEGF) and ECM proteins (Col IV and fibronectin). CONCLUSION Our findings enlighten the molecular mechanism of SC in DN by targeting the AGEs-RAGE-NF-κB signaling pathway, inflammatory responses, and ECM accumulation. Hence, the study validates the protective role of SC formulations and signifies its novel potential for treating DN.
Collapse
Affiliation(s)
- Mayura M Apte
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| | - Ekta Khattar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India.
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) (SIU), Lavale, Pune, Maharashtra State, India.
| |
Collapse
|
7
|
Lai SWT, Bhattacharya S, Lopez Gonzalez EDJ, Shuck SC. Methylglyoxal-Derived Nucleoside Adducts Drive Vascular Dysfunction in a RAGE-Dependent Manner. Antioxidants (Basel) 2024; 13:85. [PMID: 38247509 PMCID: PMC10812505 DOI: 10.3390/antiox13010085] [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: 12/07/2023] [Revised: 12/29/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Diabetic kidney disease (DKD) is a leading cause of death in patients with diabetes. An early precursor to DKD is endothelial cell dysfunction (ECD), which often precedes and exacerbates vascular disease progression. We previously discovered that covalent adducts formed on DNA, RNA, and proteins by the reactive metabolic by-product methylglyoxal (MG) predict DKD risk in patients with type 1 diabetes up to 16 years pre-diagnosis. However, the mechanisms by which MG adducts contribute to vascular disease onset and progression remain unclear. Here, we report that the most predominant MG-induced nucleoside adducts, N2-(1-carboxyethyl)-deoxyguanosine (CEdG) and N2-(1-carboxyethyl)-guanosine (CEG), drive endothelial dysfunction. Following CEdG or CEG exposure, primary human umbilical vein endothelial cells (HUVECs) undergo endothelial dysfunction, resulting in enhanced monocyte adhesion, increased reactive oxygen species production, endothelial permeability, impaired endothelial homeostasis, and exhibit a dysfunctional transcriptomic signature. These effects were discovered to be mediated through the receptor for advanced glycation end products (RAGE), as an inhibitor for intracellular RAGE signaling diminished these dysfunctional phenotypes. Therefore, we found that not only are MG adducts biomarkers for DKD, but that they may also have a role as potential drivers of vascular disease onset and progression and a new therapeutic modality.
Collapse
Affiliation(s)
- Seigmund Wai Tsuen Lai
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Edwin De Jesus Lopez Gonzalez
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| | - Sarah C. Shuck
- Department of Diabetes and Cancer Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA; (S.W.T.L.); (E.D.J.L.G.)
| |
Collapse
|
8
|
Garza-Campos A, Prieto-Correa JR, Domínguez-Rosales JA, Hernández-Nazará ZH. Implications of receptor for advanced glycation end products for progression from obesity to diabetes and from diabetes to cancer. World J Diabetes 2023; 14:977-994. [PMID: 37547586 PMCID: PMC10401444 DOI: 10.4239/wjd.v14.i7.977] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/31/2023] [Accepted: 04/17/2023] [Indexed: 07/12/2023] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are chronic pathologies with a high incidence worldwide. They share some pathological mechanisms, including hyperinsulinemia, the production and release of hormones, and hyperglycemia. The above, over time, affects other systems of the human body by causing tissue hypoxia, low-grade inflammation, and oxidative stress, which lay the pathophysiological groundwork for cancer. The leading causes of death globally are T2DM and cancer. Other main alterations of this pathological triad include the accumulation of advanced glycation end products and the release of endogenous alarmins due to cell death (i.e., damage-associated molecular patterns) such as the intracellular proteins high-mobility group box protein 1 and protein S100 that bind to the receptor for advanced glycation products (RAGE) - a multiligand receptor involved in inflammatory and metabolic and neoplastic processes. This review analyzes the latest advanced reports on the role of RAGE in the development of obesity, T2DM, and cancer, with an aim to understand the intracellular signaling mechanisms linked with cancer initiation. This review also explores inflammation, oxidative stress, hypoxia, cellular senescence, RAGE ligands, tumor microenvironment changes, and the “cancer hallmarks” of the leading tumors associated with T2DM. The assimilation of this information could aid in the development of diagnostic and therapeutic approaches to lower the morbidity and mortality associated with these diseases.
Collapse
Affiliation(s)
- Andrea Garza-Campos
- Programa de Doctorado en Ciencias en Biología Molecular en Medicina, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - José Roberto Prieto-Correa
- Programa de Doctorado en Ciencias en Biología Molecular en Medicina, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - José Alfredo Domínguez-Rosales
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Zamira Helena Hernández-Nazará
- Departamento de Biología Molecular y Genómica, Instituto de Investigación en Enfermedades Crónico-Degenerativas, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
9
|
Shu LZ, Ding YD, Xue QM, Cai W, Deng H. Direct and indirect effects of pathogenic bacteria on the integrity of intestinal barrier. Therap Adv Gastroenterol 2023; 16:17562848231176427. [PMID: 37274298 PMCID: PMC10233627 DOI: 10.1177/17562848231176427] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/01/2023] [Indexed: 06/06/2023] Open
Abstract
Bacterial translocation is a pathological process involving migration of pathogenic bacteria across the intestinal barrier to enter the systemic circulation and gain access to distant organs. This phenomenon has been linked to a diverse range of diseases including inflammatory bowel disease, pancreatitis, and cancer. The intestinal barrier is an innate structure that maintains intestinal homeostasis. Pathogenic infections and dysbiosis can disrupt the integrity of the intestinal barrier, increasing its permeability, and thereby facilitating pathogen translocation. As translocation represents an essential step in pathogenesis, a clear understanding of how barrier integrity is disrupted and how this disruption facilitates bacterial translocation could identify new routes to effective prophylaxis and therapy. In this comprehensive review, we provide an in-depth analysis of bacterial translocation and intestinal barrier function. We discuss currently understood mechanisms of bacterial-enterocyte interactions, with a focus on tight junctions and endocytosis. We also discuss the emerging concept of bidirectional communication between the intestinal microbiota and other body systems. The intestinal tract has established 'axes' with various organs. Among our regulatory systems, the nervous, immune, and endocrine systems have been shown to play pivotal roles in barrier regulation. A mechanistic understanding of intestinal barrier regulation is crucial for the development of personalized management strategies for patients with bacterial translocation-related disorders. Advancing our knowledge of barrier regulation will pave the way for future research in this field and novel clinical intervention strategies.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Yi-Dan Ding
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Qing-Ming Xue
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
| | - Wei Cai
- Medical College, Nanchang University, Nanchang,
Jiangxi Province, China
- Department of Pathology, the Fourth Affiliated
Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated
Hospital of Nanchang University, No. 133 South Guangchang Road, Nanchang
330003, Jiangxi Province, China
- Tumor Immunology Institute, Nanchang
University, Nanchang, China
| |
Collapse
|
10
|
Yan L, Li Y, Tan T, Qi J, Fang J, Guo H, Ren Z, Gou L, Geng Y, Cui H, Shen L, Yu S, Wang Z, Zuo Z. RAGE-TLR4 Crosstalk Is the Key Mechanism by Which High Glucose Enhances the Lipopolysaccharide-Induced Inflammatory Response in Primary Bovine Alveolar Macrophages. Int J Mol Sci 2023; 24:ijms24087007. [PMID: 37108174 PMCID: PMC10138623 DOI: 10.3390/ijms24087007] [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: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The receptor of advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4) are important receptors for inflammatory responses induced by high glucose (HG) and lipopolysaccharide (LPS) and show crosstalk phenomena in inflammatory responses. However, it is unknown whether RAGE and TLR4 can influence each other's expression through a crosstalk mechanism and whether the RAGE-TLR4 crosstalk related to the molecular mechanism of HG enhances the LPS-induced inflammatory response. In this study, the implications of LPS with multiple concentrations (0, 1, 5, and 10 μg/mL) at various treatment times (0, 3, 6, 12, and 24 h) in primary bovine alveolar macrophages (BAMs) were explored. The results showed that a 5 μg/mL LPS treatment at 12 h had the most significant increment on the pro-inflammatory cytokine interleukin 1β (IL-1β), IL-6, and tumor necrosis factor (TNF)-α levels in BAMs (p < 0.05) and that the levels of TLR4, RAGE, MyD88, and NF-κB p65 mRNA and protein expression were upregulated (p < 0.05). Then, the effect of LPS (5 μg/mL) and HG (25.5 mM) co-treatment in BAMs was explored. The results further showed that HG significantly enhanced the release of IL-1β, IL-6, and TNF-α caused by LPS in the supernatant (p < 0.01) and significantly increased the levels of RAGE, TLR4, MyD88, and NF-κB p65 mRNA and protein expression (p < 0.01). Pretreatment with FPS-ZM1 and TAK-242, the inhibitors of RAGE and TLR4, significantly alleviated the HG + LPS-induced increment of RAGE, TLR4, MyD88, and NF-κB p65 mRNA and protein expression in the presence of HG and LPS (p < 0.01). This study showed that RAGE and TLR4 affect each other's expression through crosstalk during the combined usage of HG and LPS and synergistically activate the MyD88/NF-κB signaling pathway to promote the release of pro-inflammatory cytokines in BAMs.
Collapse
Affiliation(s)
- Longfei Yan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Yanran Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Tianyu Tan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Jiancheng Qi
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Liping Gou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Yi Geng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Hengmin Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| | - Zhisheng Wang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611134, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611134, China
| |
Collapse
|
11
|
Ni C, Wu G, Miao T, Xu J. Wnt4 prevents apoptosis and inflammation of dental pulp cells induced by LPS by inhibiting the IKK/NF‑κB pathway. Exp Ther Med 2022; 25:75. [PMID: 36684653 PMCID: PMC9842946 DOI: 10.3892/etm.2022.11774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022] Open
Abstract
Wnt4 has been shown to promote the recovery of odontogenic differentiation of dental pulp stem cells under inflammatory conditions, but its role in inflammation and apoptosis of pulpitis remains to be elucidated. Lipopolysaccharide (LPS) (10 µg/ml) was applied to treat the human dental pulp cells (HDPCs) for 24 h. Western blotting measured the expressions of inflammatory cytokines and apoptosis-related proteins. Cell apoptosis was measured by flow cytometry. The level of Wnt4 was evaluated by reverse transcription-quantitative PCR and western blotting. The results indicated that LPS could promote inflammatory response and apoptosis in HDPCs and downregulated Wnt4 expression was found in LPS-HDPCs. Overexpression of Wnt4 ameliorated cell inflammatory response and apoptosis, presented by reduced expressions of IL-8, IL-6, TNF-α, IL-1β, Bax, cleaved-caspase 3 and enhanced Bcl-2 expression as well as decreased apoptosis rate. Moreover, overexpression of Wnt4 reduced the phosphorylation levels of IKK2, IκBα and p65 proteins upregulated by LPS. Finally, overexpression of IKK2 reversed the effects of Wnt4 on inflammation and apoptosis of LPS-HDPCs and NF-κB inhibitor reversed the effect of IKK2 overexpression in LPS-HDPCs. Wnt4 inhibited LPS-triggered inflammation and apoptosis in HDPCs via regulating the IKK/NF-κB signaling pathway, which provided a new viewpoint for understanding the pathological mechanism of pulpitis.
Collapse
Affiliation(s)
- Chengli Ni
- College of Stomatology, Anhui Medical College, Hefei, Anhui 230601, P.R. China,Correspondence to: Ms. Chengli Ni, College of Stomatology, Anhui Medical College, 632 Furong Road, Hefei, Anhui 230601, P.R. China
| | - Gang Wu
- Shanghai Smartee Denti-Technology Co., Ltd., Shanghai 200120, P.R. China
| | - Tingting Miao
- College of Stomatology, Anhui Medical College, Hefei, Anhui 230601, P.R. China
| | - Jianguang Xu
- Key Laboratory of Oral Disease Research of Anhui Province, Department of Orthodontics, Stomatologic Hospital and College, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
12
|
Versele R, Sevin E, Gosselet F, Fenart L, Candela P. TNF-α and IL-1β Modulate Blood-Brain Barrier Permeability and Decrease Amyloid-β Peptide Efflux in a Human Blood-Brain Barrier Model. Int J Mol Sci 2022; 23:ijms231810235. [PMID: 36142143 PMCID: PMC9499506 DOI: 10.3390/ijms231810235] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The blood-brain barrier (BBB) is a selective barrier and a functional gatekeeper for the central nervous system (CNS), essential for maintaining brain homeostasis. The BBB is composed of specialized brain endothelial cells (BECs) lining the brain capillaries. The tight junctions formed by BECs regulate paracellular transport, whereas transcellular transport is regulated by specialized transporters, pumps and receptors. Cytokine-induced neuroinflammation, such as the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), appear to play a role in BBB dysfunction and contribute to the progression of Alzheimer’s disease (AD) by contributing to amyloid-β (Aβ) peptide accumulation. Here, we investigated whether TNF-α and IL-1β modulate the permeability of the BBB and alter Aβ peptide transport across BECs. We used a human BBB in vitro model based on the use of brain-like endothelial cells (BLECs) obtained from endothelial cells derived from CD34+ stem cells cocultivated with brain pericytes. We demonstrated that TNF-α and IL-1β differentially induced changes in BLECs’ permeability by inducing alterations in the organization of junctional complexes as well as in transcelluar trafficking. Further, TNF-α and IL-1β act directly on BLECs by decreasing LRP1 and BCRP protein expression as well as the specific efflux of Aβ peptide. These results provide mechanisms by which CNS inflammation might modulate BBB permeability and promote Aβ peptide accumulation. A future therapeutic intervention targeting vascular inflammation at the BBB may have the therapeutic potential to slow down the progression of AD.
Collapse
Affiliation(s)
- Romain Versele
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Laurence Fenart
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Pietra Candela
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
- Correspondence:
| |
Collapse
|
13
|
Besnier E, Brunel V, Thill C, Leprêtre P, Bellien J, Demailly Z, Renet S, Tamion F, Clavier T. Soluble RAGE as a Prognostic Marker of Worsening in Patients Admitted to the ICU for COVID-19 Pneumonia: A Prospective Cohort Study. J Clin Med 2022; 11:4571. [PMID: 35956186 PMCID: PMC9369719 DOI: 10.3390/jcm11154571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/20/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Background: The different waves of SARS-CoV-2 infection have strained hospital resources and, notably, intensive care units (ICUs). Identifying patients at risk of developing a critical condition is essential to correctly refer patients to the appropriate structure and to spare limited resources. The soluble form of RAGE (sRAGE), the endoplasmic stress response and its surrogates, GRP78 and VEGF-A, may be interesting markers. Methods: This was a prospective monocenter cohort study of adult patients admitted to the ICU for severe COVID-19 pneumonia. The plasma levels of sRAGE, GRP78 and VEGF-A were measured within the first 24 h. Patients were classified as critical if they further needed vasopressor therapy, renal replacement therapy, or invasive mechanical ventilation, or died during their ICU stay, and were otherwise classified as not critical. Results: A total of 98 patients were included and 39 developed a critical condition. Critical patients presented higher sRAGE (626 [450−1043] vs. 227 [137−404] pg/mL, p < 0.0001), interleukin-6 (43 [15−112] vs. 11 [5−20] pg/mL, p < 0.0001), troponin T (17 [9−39] vs. 10 [6−18] pg/mL, p = 0.003) and NT-pro-BNP (321 [118−446] vs. 169 [63−366] pg/mL, p = 0.009) plasma levels. No difference was observed for VEGF-A and GRP78. The variables independently associated with worsening in the ICU were sRAGE (1.03 [1.01−1.05] per 10 pg/mL) and age (1.7 [1.2−2.4] per 5 years). An sRAGE value of 449.5 pg/mL predicted worsening with a sensitivity of 77% and a specificity of 80%. Conclusion: sRAGE may allow the identification of patients at risk of developing a critical form of COVID-19 pneumonia, and thus may be useful to correctly refer patients to the appropriate structure of care.
Collapse
Affiliation(s)
- Emmanuel Besnier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
- Rouen University Hospital, INSERM CIC-CRB 1404, F-76000 Rouen, France
| | - Valéry Brunel
- Department of General Biochemistry, Rouen University Hospital, F-76000 Rouen, France
| | - Caroline Thill
- Department of Biostatistics, Rouen University Hospital, F-76000 Rouen, France
| | - Perrine Leprêtre
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Jérémy Bellien
- Rouen University Hospital, INSERM CIC-CRB 1404, F-76000 Rouen, France
- Department of Pharmacology, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Zoe Demailly
- Medical Intensive Care Unit, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Sylvanie Renet
- UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Fabienne Tamion
- Medical Intensive Care Unit, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| | - Thomas Clavier
- Department of Anesthesiology and Critical Care, Rouen University Hospital, UNIROUEN, INSERM U1096, Normandie Université, F-76000 Rouen, France
| |
Collapse
|
14
|
Kim HS, Kim S, Shin SJ, Park YH, Nam Y, Kim CW, Lee KW, Kim SM, Jung ID, Yang HD, Park YM, Moon M. Gram-negative bacteria and their lipopolysaccharides in Alzheimer's disease: pathologic roles and therapeutic implications. Transl Neurodegener 2021; 10:49. [PMID: 34876226 PMCID: PMC8650380 DOI: 10.1186/s40035-021-00273-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most serious age-related neurodegenerative disease and causes destructive and irreversible cognitive decline. Failures in the development of therapeutics targeting amyloid-β (Aβ) and tau, principal proteins inducing pathology in AD, suggest a paradigm shift towards the development of new therapeutic targets. The gram-negative bacteria and lipopolysaccharides (LPS) are attractive new targets for AD treatment. Surprisingly, an altered distribution of gram-negative bacteria and their LPS has been reported in AD patients. Moreover, gram-negative bacteria and their LPS have been shown to affect a variety of AD-related pathologies, such as Aβ homeostasis, tau pathology, neuroinflammation, and neurodegeneration. Moreover, therapeutic approaches targeting gram-negative bacteria or gram-negative bacterial molecules have significantly alleviated AD-related pathology and cognitive dysfunction. Despite multiple evidence showing that the gram-negative bacteria and their LPS play a crucial role in AD pathogenesis, the pathogenic mechanisms of gram-negative bacteria and their LPS have not been clarified. Here, we summarize the roles and pathomechanisms of gram-negative bacteria and LPS in AD. Furthermore, we discuss the possibility of using gram-negative bacteria and gram-negative bacterial molecules as novel therapeutic targets and new pathological characteristics for AD.
Collapse
Affiliation(s)
- Hyeon Soo Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Chae Won Kim
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Kang Won Lee
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sung-Min Kim
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea
| | - In Duk Jung
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea
| | - Hyun Duk Yang
- Harvard Neurology Clinic, 294 Gwanggyojungang-ro, Suji-gu, Yongin, 16943, Republic of Korea.
| | - Yeong-Min Park
- Dandi Bioscience Inc, 6th Floor of Real Company Building, 66, Achasan-ro, Sungdong-gu, Seoul, Republic of Korea.
- Department of Immunology, School of Medicine, Konkuk University, 268, Chungwondaero, Chungju-si, Chungcheongbuk-do, Republic of Korea.
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea.
- Research Institute for Dementia Science, Konyang University, Daejeon, 35365, Republic of Korea.
| |
Collapse
|
15
|
The Role of HMGB1 in Traumatic Brain Injury-Bridging the Gap Between the Laboratory and Clinical Studies. Curr Neurol Neurosci Rep 2021; 21:75. [PMID: 34870759 DOI: 10.1007/s11910-021-01158-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Traumatic brain injury (TBI) is amongst the leading causes of mortality and morbidity worldwide. However, several pharmacological strategies in the clinical setting remain unsuccessful. Mounting evidence implicates High Mobility Group Box protein 1 (HMGB1) as a unique alternative target following brain injury. Herein, we discuss current understanding of HMGB1 in TBI and obstacles to clinical translation. RECENT FINDINGS HMGB1 plays a pivotal role as a 'master-switch' of neuro-inflammation following injury and in the regulation of neurogenesis during normal development. Animal models point towards the involvement of HMGB1 signalling in prolonged activation of glial cells and widespread neuronal death. Early experimental studies demonstrate positive effects of HMGB1 antagonism on both immunohistochemical and neuro-behavioural parameters following injury. Raised serum/CSF HMGB1 in humans is associated with poor outcomes post-TBI. HMGB1 is a promising therapeutic target post-TBI. However, further studies elucidating receptor, cell, isoform, and temporal effects are required prior to clinical translation.
Collapse
|
16
|
Effect of Advanced Glycation End-Products and Excessive Calorie Intake on Diet-Induced Chronic Low-Grade Inflammation Biomarkers in Murine Models. Nutrients 2021; 13:nu13093091. [PMID: 34578967 PMCID: PMC8468789 DOI: 10.3390/nu13093091] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/29/2022] Open
Abstract
Chronic Low-Grade Inflammation (CLGI) is a non-overt inflammatory state characterized by a continuous activation of inflammation mediators associated with metabolic diseases. It has been linked to the overconsumption of Advanced Glycation End-Products (AGEs), and/or macronutrients which lead to an increase in local and systemic pro-inflammatory biomarkers in humans and animal models. This review provides a summary of research into biomarkers of diet-induced CLGI in murine models, with a focus on AGEs and obesogenic diets, and presents the physiological effects described in the literature. Diet-induced CLGI is associated with metabolic endotoxemia, and/or gut microbiota remodeling in rodents. The mechanisms identified so far are centered on pro-inflammatory axes such as the interaction between AGEs and their main receptor AGEs (RAGE) or increased levels of lipopolysaccharide. The use of murine models has helped to elucidate the local and systemic expression of CLGI mediators. These models have enabled significant advances in identification of diet-induced CLGI biomarkers and resultant physiological effects. Some limitations on the translational (murine → humans) use of biomarkers may arise, but murine models have greatly facilitated the testing of specific dietary components. However, there remains a lack of information at the whole-organism level of organization, as well as a lack of consensus on the best biomarker for use in CLGI studies and recommendations as to future research conclude this review.
Collapse
|
17
|
Yuan JN, Hong Y, Ma ZL, Pang RP, Lei QQ, Lv XF, Zhou JG, Huang H, Zhang TT. MiR-302a Limits Vascular Inflammation by Suppressing Nuclear Factor-κ B Pathway in Endothelial Cells. Front Cell Dev Biol 2021; 9:682574. [PMID: 34409030 PMCID: PMC8365611 DOI: 10.3389/fcell.2021.682574] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
The inflammatory response of endothelial cells accelerates various vascular diseases. MicroRNAs (miRNAs) participate in diverse cellular processes during inflammation. In the present study, we found that miR-302a is an effective suppressor of vascular inflammation in endothelial cells. It was revealed that miR-302a exhibited a lower level in a lipopolysaccharide (LPS)-induced mouse model and in patients with vascular inflammatory disease. Genetic haploinsufficiency of miR-302 aggravated the LPS-induced vascular inflammatory response in mice, and overexpression of miR-302a attenuated vascular inflammation in mice. Furthermore, overexpression of miR-302a inhibited the synthesis and secretion of adhesion factors in endothelial cells, and suppressed the adhesion of monocytes to endothelium. In the study of molecular mechanism, we found that miR-302a relieved vascular inflammation mainly by regulating the nuclear factor kappa-B (NF-κB) pathway in endothelial cells. The results showed that interleukin-1 receptor-associated kinase4 (IRAK4) and zinc finger protein 91 (ZFP91) were the binding targets of miR-302a. MiR-302a prevented the nuclear translocation of NF-κB by inhibiting phosphorylation of IκB kinase complex β (IKKβ) and inhibitors of κBα (IκBα) via targeting IRAK4. In addition, miR-302a downregulated the expression of NF-κB by directly binding with ZFP91. These findings indicate that miR-302a negatively regulates inflammatory responses in the endothelium via the NF-κB pathway and it may be a novel target for relieving vascular inflammation.
Collapse
Affiliation(s)
- Jia-Ni Yuan
- Program of Cardiovascular Research, The Eighth Affiliated Hospital, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yu Hong
- Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhuo-Lin Ma
- Department of Physiology, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China
| | - Rui-Ping Pang
- Department of Physiology, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing-Qing Lei
- Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Fei Lv
- Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jia-Guo Zhou
- Program of Cardiovascular Research, The Eighth Affiliated Hospital, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Program of Cardiovascular Research, The Eighth Affiliated Hospital, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ting-Ting Zhang
- Program of Cardiovascular Research, The Eighth Affiliated Hospital, Zhongshan School Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Pharmacology, and Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Molecular Characteristics of RAGE and Advances in Small-Molecule Inhibitors. Int J Mol Sci 2021; 22:ijms22136904. [PMID: 34199060 PMCID: PMC8268101 DOI: 10.3390/ijms22136904] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
Receptor for advanced glycation end-products (RAGE) is a member of the immunoglobulin superfamily. RAGE binds and mediates cellular responses to a range of DAMPs (damage-associated molecular pattern molecules), such as AGEs, HMGB1, and S100/calgranulins, and as an innate immune sensor, can recognize microbial PAMPs (pathogen-associated molecular pattern molecules), including bacterial LPS, bacterial DNA, and viral and parasitic proteins. RAGE and its ligands stimulate the activations of diverse pathways, such as p38MAPK, ERK1/2, Cdc42/Rac, and JNK, and trigger cascades of diverse signaling events that are involved in a wide spectrum of diseases, including diabetes mellitus, inflammatory, vascular and neurodegenerative diseases, atherothrombosis, and cancer. Thus, the targeted inhibition of RAGE or its ligands is considered an important strategy for the treatment of cancer and chronic inflammatory diseases.
Collapse
|
19
|
Li T, Luo Q, He L, Li D, Li Q, Wang C, Xie J, Yi C. Interferon Regulatory Factor-2 Binding Protein 2 Ameliorates Sepsis-Induced Cardiomyopathy via AMPK-Mediated Anti-Inflammation and Anti-Apoptosis. Inflammation 2021; 43:1464-1475. [PMID: 32239393 DOI: 10.1007/s10753-020-01224-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cardiomyopathy commonly occurs after sepsis and is closely associated with high mortality in clinic. Interferon regulatory factor-2 binding protein 2 (IRF2BP2) has been identified as a negative regulator of inflammation, but its role in septic cardiomyopathy is unknown. The current study aims to illuminate the regulatory function of IRF2BP2 on sepsis-induced cardiomyopathy and to explore the underlying mechanisms. Protein expression of IRF2BP2 in response to sepsis-induced cardiomyopathy was examined in the heart of mice challenged by LPS intraperitoneal injection. AAV9-delivered IRF2BP2 overexpression in the heart was applied to evaluate the regulatory role of IRF2BP2 in sepsis-induced myocardial depression, inflammatory response, and cell death. The molecular mechanisms underlying IRF2BP2-regulated cardiomyopathy were explored using western blot screening assay. Primary cardiomyocytes have been isolated to further confirm the role and mechanism of IRF2BP2 during septic cardiomyopathy. IRF2BP2 expression was dramatically increased in the heart of mice after LPS administration. AAV9-mediated IRF2BP2 overexpression significantly improved sepsis-induced cardiac dysfunction, inhibited inflammatory cell infiltration and cytokine production, and blocked cell death after LPS treatment. Mechanistically, IRF2BP2 activated AMPK signaling in cardiomyocytes, while inhibiting AMPK activation largely reversed IRF2BP2-benefited inflammatory suppression and cell survival. These findings clearly demonstrated that IRF2BP2 is a potent suppressor of sepsis-induced myocardial depression and related heart impairment. Targeting IRF2BP2 represents a promising therapeutic strategy for septic cardiomyopathy.
Collapse
Affiliation(s)
- Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Qiang Luo
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Li He
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Da Li
- Department of Pharmacy, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Qingnian Li
- Department of Intensive Care Unit, Tongji Hospital, Tongji Medical College of Huazhong, University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Chuntao Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Jie Xie
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China
| | - Chengla Yi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang avenue, Wuhan, 430030, China.
| |
Collapse
|
20
|
Abouelasrar Salama S, Gouwy M, Van Damme J, Struyf S. The turning away of serum amyloid A biological activities and receptor usage. Immunology 2021; 163:115-127. [PMID: 33315264 PMCID: PMC8114209 DOI: 10.1111/imm.13295] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022] Open
Abstract
Serum amyloid A (SAA) is an acute-phase protein (APP) to which multiple immunological functions have been attributed. Regardless, the true biological role of SAA remains poorly understood. SAA is remarkably conserved in mammalian evolution, thereby suggesting an important biological function. Since its discovery in the 1970s, the majority of researchers have investigated SAA using recombinant forms made available through bacterial expression. Nevertheless, recent studies indicate that these recombinant forms of SAA are unreliable. Indeed, commercial SAA variants have been shown to be contaminated with bacterial products including lipopolysaccharides and lipoproteins. As such, biological activities and receptor usage (TLR2, TLR4) revealed through the use of commercial SAA variants may not reflect the inherent nature of this APP. Within this review, we discuss the biological effects of SAA that have been demonstrated through more solid experimental approaches. SAA takes part in the innate immune response via the recruitment of leucocytes and executes, through pathogen recognition, antimicrobial activity. Knockout animal models implicate SAA in a range of functions, such as regulation of T-cell-mediated responses and monopoiesis. Moreover, through its structural motifs, not only does SAA function as an extracellular matrix protein, but it also binds extracellular matrix proteins. Finally, we here also provide an overview of definite SAA receptor-mediated functions and highlight those that are yet to be validated. The role of FPR2 in SAA-mediated leucocyte recruitment has been confirmed; nevertheless, SAA has been linked to a range of other receptors including CD36, SR-BI/II, RAGE and P2RX7.
Collapse
Affiliation(s)
- Sara Abouelasrar Salama
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven, Belgium
| |
Collapse
|
21
|
Zheng Y, Wang Y, Zhu X, Diao Y, Chen Y, Chen G. Circ_0038467 regulates lipopolysaccharide-mediated cell proliferation, apoptosis, and inflammatory response by miR-195-5p/TLR4 axis through NF-κB pathway in MRC-5 cells. Biosci Biotechnol Biochem 2021; 85:1639-1649. [PMID: 34021567 DOI: 10.1093/bbb/zbab092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Infantile pneumonia (IP) is an acute lower respiratory infection and brings a heavy burden to children health. Circular RNAs (circRNAs) participate in the regulation of pneumonia process. In this research, the effects of circ_0038467 in regulating lipopolysaccharide (LPS)-induced cell injury and underlying mechanism were revealed. Results showed that circ_0038467 expression and TLR4 protein level were upregulated, while miR-195-5p expression was downregulated in LPS-induced MRC-5 cells. Circ_0038467 silencing restored LPS-mediated inhibition on cell proliferation and promotion on apoptosis and inflammatory response. Additionally, circ_0038467 acted as a sponge of miR-195-5p, which was further revealed to target TLR4. MiR-195-5p inhibitor reversed circ_0038467 silencing-mediated influences under LPS treatment. Furthermore, LPS-activated NF-κB pathway was partly blocked by circ_0038467 silencing, which was restrained by TLR4 overexpression. Circ_0038467 silencing protected MRC-5 cells from LPS-induced injury by miR-195-5p/TLR4 axis through NF-κB pathway, providing a theoretical basis for circRNA-directed IP therapy.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuanyuan Wang
- Department of Respiratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiuli Zhu
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuqiao Diao
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuqin Chen
- Department of Respiratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Gang Chen
- Department of Respiratory, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
22
|
Follacchio GA, Manganelli V, Monteleone F, Sorice M, Garofalo T, Liberatore M. HMGB1 expression in leukocytes as a biomarker of cellular damage induced by [ 99mTc]Tc-HMPAO-labelling procedure: A quality control study. Nucl Med Biol 2021; 96-97:94-100. [PMID: 33864964 DOI: 10.1016/j.nucmedbio.2021.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE Autologous White Blood Cells (WBC) scintigraphy is based on a multi-step sequence of cell separation and radiolabelling. Besides in vivo imaging quality control, no molecular tool is available to evaluate WBC damage secondary to cell manipulation. High Mobility Group Box 1 (HMGB1) is a protein of the alarmins family, secreted by innate immune cells and released from the nucleus of damaged cells following different types of injury. Aim of this study was to evaluate HMGB1 levels in WBC cytosolic extracts (CE) before and after [99mTc]Tc-HMPAO labelling procedure, as a biomarker of induced WBC damage. PROCEDURES Patients with suspect of prosthetic joint infection were prospectively enrolled. HMGB1 levels were evaluated by immunoblotting analysis in plasma (t0), and in WBC-CE before (t1) and after (t2) [99mTc]Tc-HMPAO labelling. Blood samples from healthy subjects were evaluated under the same procedure. RESULTS Twenty consecutive patients referred for WBC scintigraphy and ten controls were enrolled. HMGB1 levels were significantly upregulated both in plasma (t0) and in circulating WBC-CE (t1) from patients compared to controls (p < 0.0001). Otherwise, WBC-CE from [99mTc]Tc-HMPAO-labelled leukocyte concentrate (t2) did not show significant changes in HMGB1 levels compared to the cold leukocyte sample (t1). CONCLUSIONS The evaluation of HMGB1 levels in WBC-CE from each subject after radiolabelling with [99mTc]Tc-HMPAO did not show significant changes compared to the cold cellular sample. These results further prove the reliability of [99mTc]Tc-HMPAO leukocyte radiolabelling procedure in terms of cell viability and suggest that the monitoring of this alarmin may represent a specific tool to evaluate a secondary damage of WBC induced by radiolabelling procedure. In addition, significant upregulation of HMGB1 levels was found in WBC-CE and in plasma from patients with suspect of PJI - compared to healthy donors - reasonably related to their underlying inflammatory/infective condition.
Collapse
Affiliation(s)
- Giulia Anna Follacchio
- Nuclear Medicine Unit, Department of Radiology, Oncology and Human Pathology, "Sapienza" University of Rome, Italy; Molecular Medicine PhD Program, Department of Molecular Medicine, "Sapienza" University of Rome, Italy.
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Francesco Monteleone
- Nuclear Medicine Unit, Department of Radiology, Oncology and Human Pathology, "Sapienza" University of Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, Italy
| | - Mauro Liberatore
- Nuclear Medicine Unit, Department of Radiology, Oncology and Human Pathology, "Sapienza" University of Rome, Italy
| |
Collapse
|
23
|
Wong E, Xu F, Joffre J, Nguyen N, Wilhelmsen K, Hellman J. ERK1/2 Has Divergent Roles in LPS-Induced Microvascular Endothelial Cell Cytokine Production and Permeability. Shock 2021; 55:349-356. [PMID: 32826812 PMCID: PMC8139579 DOI: 10.1097/shk.0000000000001639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Endothelial cells play a major role in inflammatory responses to infection and sterile injury. Endothelial cells express Toll-like receptor 4 (TLR4) and are activated by LPS to express inflammatory cytokines/chemokines, and to undergo functional changes, including increased permeability. The extracellular signal-regulated kinase 1/2 (ERK1/2) mediates pro-inflammatory signaling in monocytes and macrophages, but the role of ERK1/2 in LPS-induced activation of microvascular endothelial cells has not been defined. We therefore studied the role of ERK1/2 in LPS-induced inflammatory activation and permeability of primary human lung microvascular endothelial cells (HMVEC). Inhibition of ERK1/2 augmented LPS-induced IL-6 and vascular cell adhesion protein (VCAM-1) production by HMVEC. ERK1/2 siRNA knockdown also augmented IL-6 production by LPS-treated HMVEC. Conversely, ERK1/2 inhibition abrogated permeability and restored cell-cell junctions of LPS-treated HMVEC. Consistent with the previously described pro-inflammatory role for ERK1/2 in leukocytes, inhibition of ERK1/2 reduced LPS-induced cytokine/chemokine production by primary human monocytes. Our study identifies a complex role for ERK1/2 in TLR4-activation of HMVEC, independent of myeloid differentiation primary response gene (MyD88) and TIR domain-containing adaptor inducing IFN-β (TRIF) signaling pathways. The activation of ERK1/2 limits LPS-induced IL-6 production by HMVEC, while at the same time promoting HMVEC permeability. Conversely, ERK1/2 activation promotes IL-6 production by human monocytes. Our results suggest that ERK1/2 may play an important role in the nuanced regulation of endothelial cell inflammation and vascular permeability in sepsis and injury.
Collapse
Affiliation(s)
- Erika Wong
- Department of Pediatrics, Division of Critical Care, UCSF Benioff Children’s Hospital, San Francisco, California, 94143
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, 94143
| | - Fengyun Xu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, 94143
| | - Jérémie Joffre
- Medical Intensive Care Unit, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, 75571 Paris cedex 12, France
| | - Nina Nguyen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, 94143
| | - Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, 94143
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California, 94143
| |
Collapse
|
24
|
From "Leaky Gut" to Impaired Glia-Neuron Communication in Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:129-155. [PMID: 33834399 DOI: 10.1007/978-981-33-6044-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the last three decades, the robust scientific data emerged, demonstrating that the immune-inflammatory response is a fundamental component of the pathophysiology of major depressive disorder (MDD). Psychological stress and various inflammatory comorbidities contribute to such immune activation. Still, this is not uncommon that patients with depression do not have defined inflammatory comorbidities, and alternative mechanisms of immune activation need to take place. The gastrointestinal (GI) tract, along with gut-associated lymphoid tissue (GALT), constitutes the largest lymphatic organ in the human body and forms the biggest surface of contact with the external environment. It is also the most significant source of bacterial and food-derived antigenic material. There is a broad range of reciprocal interactions between the GI tract, intestinal microbiota, increased intestinal permeability, activation of immune-inflammatory response, and the CNS that has crucial implications in brain function and mental health. This intercommunication takes place within the microbiota-gut-immune-glia (MGIG) axis, and glial cells are the main orchestrator of this communication. A broad range of factors, including psychological stress, inflammation, dysbiosis, may compromise the permeability of this barrier. This leads to excessive bacterial translocation and the excessive influx of food-derived antigenic material that contributes to activation of the immune-inflammatory response and depressive psychopathology. This chapter summarizes the role of increased intestinal permeability in MDD and mechanisms of how the "leaky gut" may contribute to immune-inflammatory response in this disorder.
Collapse
|
25
|
Ullah R, Ikram M, Park TJ, Ahmad R, Saeed K, Alam SI, Rehman IU, Khan A, Khan I, Jo MG, Kim MO. Vanillic Acid, a Bioactive Phenolic Compound, Counteracts LPS-Induced Neurotoxicity by Regulating c-Jun N-Terminal Kinase in Mouse Brain. Int J Mol Sci 2020; 22:ijms22010361. [PMID: 33396372 PMCID: PMC7795830 DOI: 10.3390/ijms22010361] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
The receptor for advanced glycation end products (RAGE), a pattern recognition receptor signaling event, has been associated with several human illnesses, including neurodegenerative diseases, particularly in Alzheimer’s disease (AD). Vanillic acid (V.A), a flavoring agent, is a benzoic acid derivative having a broad range of biological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the underlying molecular mechanisms of V.A in exerting neuroprotection are not well investigated. The present study aims to explore the neuroprotective effects of V.A against lipopolysaccharides (LPS)-induced neuroinflammation, amyloidogenesis, synaptic/memory dysfunction, and neurodegeneration in mice brain. Behavioral tests and biochemical and immunofluorescence assays were applied. Our results indicated increased expression of RAGE and its downstream phospho-c-Jun n-terminal kinase (p-JNK) in the LPS-alone treated group, which was significantly reduced in the V.A + LPS co-treated group. We also found that systemic administration of LPS-injection induced glial cells (microglia and astrocytes) activation and significantly increased expression level of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB) and secretion of proinflammatory cytokines including tumor necrosis factor alpha (TNF-α), interleukin-1 β (IL1-β), and cyclooxygenase (COX-2). However, V.A + LPS co-treatment significantly inhibited the LPS-induced activation of glial cells and neuroinflammatory mediators. Moreover, we also noted that V.A treatment significantly attenuated LPS-induced increases in the expression of AD markers, such as β-site amyloid precursor protein (APP)–cleaving enzyme 1 (BACE1) and amyloid-β (Aβ). Furthermore, V.A treatment significantly reversed LPS-induced synaptic loss via enhancing the expression level of pre- and post-synaptic markers (PSD-95 and SYP), and improved memory performance in LPS-alone treated group. Taken together; we suggest that neuroprotective effects of V.A against LPS-induced neurotoxicity might be via inhibition of LPS/RAGE mediated JNK signaling pathway; and encourage future studies that V.A would be a potential neuroprotective and neurotherapeutic candidate in various neurological disorders.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Muhammad Ikram
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Tae Ju Park
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow G12OZD, UK;
| | - Riaz Ahmad
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Kamran Saeed
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Sayed Ibrar Alam
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Inayat Ur Rehman
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Amjad Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Ibrahim Khan
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Min Gi Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (R.U.); (M.I.); (R.A.); (K.S.); (S.I.A.); (I.U.R.); (A.K.); (I.K.); (M.G.J.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
26
|
A Review of Malaysian Herbal Plants and Their Active Constituents with Potential Therapeutic Applications in Sepsis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8257817. [PMID: 33193799 PMCID: PMC7641701 DOI: 10.1155/2020/8257817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Sepsis refers to organ failure due to uncontrolled body immune responses towards infection. The systemic inflammatory response triggered by pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide (LPS) from Gram-negative bacteria, is accompanied by the release of various proinflammatory mediators that can lead to organ damage. The progression to septic shock is even more life-threatening due to hypotension. Thus, sepsis is a leading cause of death and morbidity globally. However, current therapies are mainly symptomatic treatment and rely on the use of antibiotics. The lack of a specific treatment demands exploration of new drugs. Malaysian herbal plants have a long history of usage for medicinal purposes. A total of 64 Malaysian plants commonly used in the herbal industry have been published in Malaysian Herbal Monograph 2015 and Globinmed website (http://www.globinmed.com/). An extensive bibliographic search in databases such as PubMed, ScienceDirect, and Scopus revealed that seven of these plants have antisepsis properties, as evidenced by the therapeutic effect of their extracts or isolated compounds against sepsis-associated inflammatory responses or conditions in in vitro or/and in vivo studies. These include Andrographis paniculata, Zingiber officinale, Curcuma longa, Piper nigrum, Syzygium aromaticum, Momordica charantia, and Centella asiatica. Among these, Z. officinale is the most widely studied plant and seems to have the highest potential for future therapeutic applications in sepsis. Although both extracts as well as active constituents from these herbal plants have demonstrated potential antisepsis activity, the activity might be primarily contributed by the active constituent(s) from each of these plants, which are andrographolide (A. paniculata), 6-gingerol and zingerone (Z. officinale), curcumin (C. longa), piperine and pellitorine (P. nigrum), biflorin (S. aromaticum), and asiaticoside, asiatic acid, and madecassoside (C. asiatica). These active constituents have shown great antisepsis effects, and further investigations into their clinical therapeutic potential may be worthwhile.
Collapse
|
27
|
Matsumoto NM, Aoki M, Okubo Y, Kuwahara K, Eura S, Dohi T, Akaishi S, Ogawa R. Gene Expression Profile of Isolated Dermal Vascular Endothelial Cells in Keloids. Front Cell Dev Biol 2020; 8:658. [PMID: 32850798 PMCID: PMC7403211 DOI: 10.3389/fcell.2020.00658] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 07/01/2020] [Indexed: 01/01/2023] Open
Abstract
Wound healing is a complex biological process, and imbalances of various substances in the wound environment may prolong healing and lead to excessive scarring. Keloid is abnormal proliferation of scar tissue beyond the original wound margins with excessive deposition of extracellular matrix (ECM) and chronic inflammation. Despite numerous previous research efforts, the pathogenesis of keloid remains unknown. Vascular endothelial cells (VECs) are a major type of inductive cell in inflammation and fibrosis. Despite several studies on vascular morphology in keloid formation, there has been no functional analysis of the role of VECs. In the present study, we isolated living VECs from keloid tissues and investigated gene expression patterns using microarray analysis. We obtained 5 keloid tissue samples and 6 normal skin samples from patients without keloid. Immediately after excision, tissue samples were gently minced and living cells were isolated. Magnetic-activated cell sorting of VECs was performed by negative selection of fibroblasts and CD45+ cells and by positive selection of CD31+cells. After RNA extraction, gene expression analysis was performed to compare VECs isolated from keloid tissue (KVECs) with VECs from normal skin (NVECs). After cell isolation, the percentage of CD31+ cells as measured by flow cytometry ranged from 81.8%–98.6%. Principal component analysis was used to identify distinct molecular phenotypes in KVECs versus NVECs and these were divided into two subgroups. In total, 15 genes were upregulated, and 3 genes were downregulated in KVECs compared with NVECs using the t-test (< 0.05). Quantitative RT-PCR and immunohistochemistry showed 16-fold and 11-fold overexpression of SERPINA3 and LAMC2, respectively. SERPINA3 encodes the serine protease inhibitor, α1-antichymotripsin. Laminin γ2-Chain (LAMC2) is a subunit of laminin-5 that induces retraction of vascular endothelial cells and enhances vascular permeability. This is the first report of VEC isolation and gene expression analysis in keloid tissue. Our data suggest that SERPINA3 and LAMC2 upregulation in KVECs may contribute to the development of fibrosis and prolonged inflammation in keloid. Further functional investigation of these genes will help clarify the mechanisms of abnormal scar tissue proliferation.
Collapse
Affiliation(s)
- Noriko M Matsumoto
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Masayo Aoki
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Yuri Okubo
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Kosuke Kuwahara
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Shigeyoshi Eura
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Teruyuki Dohi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Satoshi Akaishi
- Department of Plastic Surgery, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Japan
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
28
|
Wu J, Deng Z, Sun M, Zhang W, Yang Y, Zeng Z, Wu J, Zhang Q, Liu Y, Chen Z, Guo X, Zhao KS, Huang Q, Chen Z. Polydatin protects against lipopolysaccharide-induced endothelial barrier disruption via SIRT3 activation. J Transl Med 2020; 100:643-656. [PMID: 31641228 DOI: 10.1038/s41374-019-0332-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/30/2019] [Accepted: 09/23/2019] [Indexed: 12/15/2022] Open
Abstract
In a previous study, we demonstrated the role of polydatin (PD) in protecting against multiple organ dysfunction in sepsis. The aim of this study is to investigate whether PD protects against lipopolysaccharide (LPS)-induced endothelial barrier disruption through SIRT3 activation and to disclose the underlying mechanisms. Wild-type mice were injected with LPS and Evans Blue assay was performed to evaluate vascular permeability. Primary human umbilical vein endothelial cells (HUVECs) were stimulated with LPS. Endothelial permeability was evaluated by transendothelial electrical resistance (TER) and FITC-dextran leakage. SIRT3 activity was determined by a Deacetylase Fluorometric kit, and protein expression level of SIRT3 was detected by western blotting. Mitochondrial function was evaluated by determination of ROS level, mitochondrial membrane potential and mPTP opening. In endotoxemic mice, PD pretreatment attenuated vascular leakage in multiple organs while SIRT3 inhibition with 3-TYP reversed the effects of PD. PD treatment in late sepsis also exhibited barrier protective effects. In HUVECs, PD alleviated LPS-induced F-actin rearrangement, cadherin-catenin complex dissociation and endothelial hyperpermeability, whereas 3-TYP or SIRT3 siRNA attenuated the protective effects of PD. PD enhanced SIRT3 deacetylase activity, and attenuated LPS-induced decrease in SIRT3 expression as well. Furthermore, gain-of-function and loss-of-function strategies also confirmed the role of SIRT3 in enhancing endothelial barrier integrity. It was further ascertained that PD enhanced SIRT3-mediated deacetylation of SOD2 and cyclophilin D (CypD), thus suppressing mitochondrial dysfunction and subsequent endothelial barrier dysfunction. In addition, it was revealed that RAGE was involved in LPS-regulated SIRT3 signaling. Our results suggest that polydatin protects against LPS-induced endothelial barrier disruption dependent on SIRT3 and can be applied as a potential therapy for sepsis.
Collapse
Affiliation(s)
- Jie Wu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Zhiya Deng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Maomao Sun
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Weijin Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.,Department of Geriatrics, Shantou Central Hospital, Shantou, 515000, China
| | - Yang Yang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Department of Critical Care Medicine, the First People's Hospital of Chenzhou, Chenzhou, 423000, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Jianhua Wu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Qin Zhang
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenfeng Chen
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Xiaohua Guo
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Ke-Seng Zhao
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China
| | - Qiaobing Huang
- Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Guangdong Provincial Key Lab of Shock and Microcirculation, Department of Pathophysiology, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
29
|
Involvement of Receptor for Advanced Glycation Endproducts in Hypertensive Disorders of Pregnancy. Int J Mol Sci 2019; 20:ijms20215462. [PMID: 31683992 PMCID: PMC6862609 DOI: 10.3390/ijms20215462] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia/hypertensive disorders of pregnancy (PE/HDP) is a serious and potentially life-threatening disease. Recently, PE/HDP has been considered to cause adipose tissue inflammation, but the detailed mechanism remains unknown. We exposed human primary cultured adipocytes with serum from PE/HDP and healthy controls for 24 h, and analyzed mRNA expression of several adipokines, cytokines, and ligands of the receptor for advanced glycation endproducts (RAGE). We found that the mRNA levels of interleukin-6 (IL-6), C-C motif chemokine ligand 2 (CCL2), high mobility group box 1 (HMGB1), and RAGE were significantly increased by the addition of PE/HDP serum. Among RAGE ligands, advanced glycation endproducts (AGE) and HMGB1 increased mRNA levels of IL-6 and CCL2 in SW872 human adipocytes and mouse 3T3-L1 cells. The introduction of small interfering RNA for RAGE (siRAGE) into SW872 cells abolished the AGE- and HMGB1-induced up-regulation of IL-6 and CCL2. In addition, lipopolysaccharide (LPS), a ligand of RAGE, increased the expression of IL-6 and CCL2 and siRAGE attenuated the LPS-induced expression of IL-6 and CCL2. These results strongly suggest that the elevated AGE, HMGB1, and LPS in pregnant women up-regulate the expression of IL-6 and CCL2 via the RAGE system, leading to systemic inflammation such as PE/HDP.
Collapse
|
30
|
Ni J, Lin M, Jin Y, Li J, Guo Y, Zhou J, Hong G, Zhao G, Lu Z. Gas6 Attenuates Sepsis-Induced Tight Junction Injury and Vascular Endothelial Hyperpermeability via the Axl/NF-κB Signaling Pathway. Front Pharmacol 2019; 10:662. [PMID: 31263416 PMCID: PMC6585310 DOI: 10.3389/fphar.2019.00662] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial functional dysregulation and barrier disruption are involved the initiation and development of sepsis. Growth arrest-specific protein 6 (Gas6), one of the endogenous ligands of TAM receptors (Tyro3, Axl, and Mertk), is confirmed to have beneficial functions in hemostasis, inflammation, and cancer growth. Here, we demonstrated the protective effects of Gas6 on multi-organ dysfunction syndrome (MODS) in sepsis and the underlying mechanisms. We investigated Gas6-ameliorated MODS by inhibiting vascular endothelial hyperpermeability in a mouse model of sepsis. Additionally, in vitro, under lipopolysaccharide (LPS) stimulation in vascular endothelial cells, Gas6 attenuated vascular endothelial hyperpermeability by reinforcing the tight junction proteins occludin, zonula occludens-1 (ZO-1), and claudin5. Furthermore, Gas6 substantially suppressed NF-κB p65 activation. In addition, blocking the Gas6 receptor, Axl, partially reduced the protective effect of Gas6 on the vascular endothelial barrier and diminished the inhibitive effect of Gas6 on NF-κB p65 activation. Taken together, this study suggests that Gas6 has a protective effect on MODS in sepsis by inhibiting the vascular endothelial hyperpermeability and alteration of tight junction and that the Axl/NF-κB signaling pathway underlies these effects.
Collapse
Affiliation(s)
- Jingjing Ni
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Miaotong Lin
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yangjie Jin
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiajia Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yayong Guo
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jindong Zhou
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guangliang Hong
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guangju Zhao
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongqiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
31
|
Manganelli V, Truglia S, Capozzi A, Alessandri C, Riitano G, Spinelli FR, Ceccarelli F, Mancuso S, Garofalo T, Longo A, Valesini G, Sorice M, Conti F, Misasi R. Alarmin HMGB1 and Soluble RAGE as New Tools to Evaluate the Risk Stratification in Patients With the Antiphospholipid Syndrome. Front Immunol 2019; 10:460. [PMID: 30923525 PMCID: PMC6426766 DOI: 10.3389/fimmu.2019.00460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antiphospholipid antibody syndrome (APS) is a systemic autoimmune disease characterized by arterial and/or venous thrombosis, pregnancy morbidity in the presence of circulating “anti-phospholipid antibodies” (aPL). One of the main target antigens of aPL is β2-glycoprotein I (β2-GPI). APS may occur as a primary syndrome or associated with Systemic Lupus Erythematosus (SLE). High Mobility Group Box 1 (HMGB1) is a nuclear non-histone protein which is secreted from different type of cells during activation and/or cell death and may act as a proinflammatory mediator through ligation to its receptors, including RAGE. There is accumulating evidence that HMGB1 contributes to the pathogenesis of inflammatory and autoimmune diseases, especially SLE. In a previous study we demonstrated increased serum levels of HMGB1 in both primary and secondary APS patients. In this work we analyzed: (i) in vitro whether anti-β2-GPI antibodies from APS patients may induce both a HMGB1 cellular relocation by activation of its putative receptor RAGE in platelets and monocytes and, (ii) ex vivo, serum levels of HMGB1/soluble RAGE (sRAGE) in APS patients and their possible correlation with clinical manifestations. Platelets and monocytes from healthy donors were incubated with affinity purified anti-β2-GPI antibodies. HMGB1 and RAGE expression were analyzed by Western Blot. Sera from 60 consecutive APS patients (primary or secondary), diagnosed according to the Sydney Classification Criteria, were enrolled. As a control, 30 matched healthy subjects were studied. Serum levels of HMGB1 and sRAGE were analyzed by Western Blot. In vitro results showed that anti-β2-GPI antibodies were able to induce RAGE activation and HMGB1 cellular relocation in both monocytes and platelets. HMGB1 and sRAGE serum levels were significantly increased in APS patients in comparison with healthy subjects (p<0.0001). Interestingly, APS patients with spontaneous recurrent abortion showed significantly higher levels of sRAGE; moreover, in APS patients a direct correlation between serum levels of HMGB1 and disease duration was detected. Our observations suggest that anti-β2-GPI antibodies may trigger RAGE activation and HMGB1 cellular relocation during APS. Monitoring these molecules serum levels may represent an useful tool to evaluate the pathogenesis and risk stratification of clinical manifestations in APS.
Collapse
Affiliation(s)
- Valeria Manganelli
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Simona Truglia
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Antonella Capozzi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Cristiano Alessandri
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Gloria Riitano
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Francesca Romana Spinelli
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Fulvia Ceccarelli
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Silvia Mancuso
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Tina Garofalo
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Agostina Longo
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Guido Valesini
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Maurizio Sorice
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| | - Fabrizio Conti
- Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Roberta Misasi
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
32
|
Lu H, Wen D, Wang X, Gan L, Du J, Sun J, Zeng L, Jiang J, Zhang A. Host genetic variants in sepsis risk: a field synopsis and meta-analysis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:26. [PMID: 30683156 PMCID: PMC6347778 DOI: 10.1186/s13054-019-2313-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/07/2019] [Indexed: 01/07/2023]
Abstract
Background Published data revealed that host genetic variants have a substantial influence on sepsis susceptibility. However, the results have been inconsistent. We aimed to systematically review the published studies and quantitatively evaluate the effects of these variants on the risk of sepsis. Methods We searched the PubMed, EMBASE, Medline, Web of Knowledge, and HuGE databases to identify studies that investigated the associations between genetic variants and sepsis risk. Then, we conducted meta-analyses of the associations for genetic variants with at least three study populations and applied the Venice criteria to assess the association result credibility. Results A literature search identified 349 eligible articles that investigated 405 variants of 172 distinct genes. We performed 204 primary and 185 subgroup meta-analyses for 76 variants of 44 genes. The results showed that 29 variants of 23 genes were significantly associated with the risk of sepsis, including 8 variants of pattern recognition receptors (PRRs), 14 variants of cytokines, one variant of an immune-related gene and 6 variants of other genes. Furthermore, the cumulative epidemiological evidence of a significant association between each variant and the risk of sepsis was classified as strong or moderate for 18 variants. For the 329 variants with fewer than three study populations, 63 variants of 48 genes have been reported to be significantly associated with the risk of sepsis in a systematic review. Conclusion We identified several genetic variants that could influence the susceptibility to sepsis by systematic review and meta-analysis. This study provides a comprehensive overview of the genetic architecture of variants involved in sepsis susceptibility and novel insight that may affect personalized targeted treatment in the future clinical management of sepsis. Electronic supplementary material The online version of this article (10.1186/s13054-019-2313-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongxiang Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China
| | - Dalin Wen
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China
| | - Xu Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China.,Department of Emergency Surgery, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Lebin Gan
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China.,Department of Emergency Surgery, The Affiliated Hospital, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China
| | - Jianhui Sun
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China.
| | - Anqiang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Changjiang Branch Road 10, Daping Street, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
33
|
Kutryb-Zajac B, Mierzejewska P, Sucajtys-Szulc E, Bulinska A, Zabielska MA, Jablonska P, Serocki M, Koszalka P, Milczarek R, Jasztal A, Bartoszewski R, Chlopicki S, Slominska EM, Smolenski RT. Inhibition of LPS-stimulated ecto-adenosine deaminase attenuates endothelial cell activation. J Mol Cell Cardiol 2019; 128:62-76. [PMID: 30641086 DOI: 10.1016/j.yjmcc.2019.01.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/23/2018] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Vascular inflammation is an important factor in the pathophysiology of cardiovascular diseases, such as atherosclerosis. Changes in the extracellular nucleotide and in particular adenosine catabolism may alter a chronic inflammation and endothelial activation. This study aimed to evaluate the relation between vascular ecto-adenosine deaminase (eADA) activity and endothelial activation in humans and to analyze the effects of LPS-mediated inflammation on this activity as well as mechanisms of its increase. Moreover, we investigated a therapeutic potential of ADA inhibition by deoxycofromycin (dCF) for endothelial activation. We demonstrated a positive correlation of vascular eADA activity and ADA1 mRNA expression with endothelial activation parameters in humans with atherosclerosis. The activation of vascular eADA was also observed under LPS stimulation in vivo along with endothelial activation, an increase in markers of inflammation and alterations in the lipid profile of a rat model. Ex vivo and in vitro studies on human specimen demonstrated that at an early stage of vascular pathology, eADA activity originated from activated endothelial cells, while at later stages also from an inflammatory infiltrate. We proposed that LPS-stimulated increase in endothelial adenosine deaminase activity could be a result of IL-6/JAK/STAT pathway activation, since the lack of IL-6 in mice was associated with lower vascular and plasma eADA activities. Furthermore, the inhibitors of JAK/STAT pathway decreased LPS-stimulated adenosine deaminase activity in endothelial cells. We demonstrated that cell surface eADA activity could be additionally regulated by transcytosis pathways, as exocytosis inhibitors including lipid raft inhibitor, methyl-β-cyclodextrin decreased LPS-induced eADA activity. This suggests that cholesterol-dependent protein externalization mediated by lipid rafts could be an important factor in the eADA increase. Moreover, endocytosis inhibitors and exocytosis activators increased this activity on the cell surface. Furthermore, the inhibition of adenosine deaminase in endothelial cells in vitro attenuated LPS-mediated IL-6 release and soluble ICAM-1 and VCAM-1 concentration in the incubation medium through the restoration of the extracellular adenosine pool and adenosine receptor-dependent pathways. This study demonstrated that the vascular endothelial eADA activity remains under control of inflammatory mediators acting through JAK/STAT pathway that could be further modified by dyslipidemic-dependent exocytosis and transcytosis pathways. Inhibition of eADA blocked endothelial activation suggesting a crucial role of this enzyme in the control of vascular inflammation. This supports the concept of eADA targeted vascular protection therapy.
Collapse
Affiliation(s)
- Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Paulina Mierzejewska
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Elzbieta Sucajtys-Szulc
- Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdansk, 7 Debinki St., 80-952 Gdansk, Poland
| | - Alicja Bulinska
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Magdalena A Zabielska
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland; Department of Physiology, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Patrycja Jablonska
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Marcin Serocki
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107 St., 80-416 Gdansk, Poland
| | - Patrycja Koszalka
- Department of Medical Biotechnology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Ryszard Milczarek
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348 Krakow, Poland
| | - Rafal Bartoszewski
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Hallera 107 St., 80-416 Gdansk, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348 Krakow, Poland
| | - Ewa M Slominska
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdansk, 1 Debinki St., 80-211 Gdansk, Poland.
| |
Collapse
|
34
|
Endothelial Protrusions in Junctional Integrity and Barrier Function. CURRENT TOPICS IN MEMBRANES 2018; 82:93-140. [PMID: 30360784 DOI: 10.1016/bs.ctm.2018.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Endothelial cells of the microcirculation form a semi-permeable diffusion barrier between the blood and tissues. This permeability of the endothelium, particularly in the capillaries and postcapillary venules, is a normal physiological function needed for blood-tissue exchange in the microcirculation. During inflammation, microvascular permeability increases dramatically and can lead to tissue edema, which in turn can lead to dysfunction of tissues and organs. The molecular mechanisms that control the barrier function of endothelial cells have been under investigation for several decades and remain an important topic due to the potential for discovery of novel therapeutic strategies to reduce edema. This review highlights current knowledge of the cellular and molecular mechanisms that lead to endothelial hyperpermeability during inflammatory conditions associated with injury and disease. This includes a discussion of recent findings demonstrating temporal protrusions by endothelial cells that may contribute to intercellular junction integrity between endothelial cells and affect the diffusion distance for solutes via the paracellular pathway.
Collapse
|
35
|
Gasparotto J, Ribeiro CT, da Rosa-Silva HT, Bortolin RC, Rabelo TK, Peixoto DO, Moreira JCF, Gelain DP. Systemic Inflammation Changes the Site of RAGE Expression from Endothelial Cells to Neurons in Different Brain Areas. Mol Neurobiol 2018; 56:3079-3089. [DOI: 10.1007/s12035-018-1291-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/31/2018] [Indexed: 12/21/2022]
|
36
|
Atorvastatin enhances endothelial adherens junctions through promoting VE-PTP gene transcription and reducing VE-cadherin-Y731 phosphorylation. Vascul Pharmacol 2018; 117:7-14. [PMID: 29894844 DOI: 10.1016/j.vph.2018.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/03/2018] [Accepted: 06/03/2018] [Indexed: 12/17/2022]
Abstract
Vascular endothelial protein tyrosine phosphatase (VE-PTP) is essential for endothelial cells (ECs) adherens junction and vascular homeostasis; however, the regulatory mechanism of VE-PTP transcription is unknown, and a drug able to promote VE-PTP expression in ECs has not yet been reported in the literature. In this study, we used human ECs as a model to explore small molecule compounds able to promote VE-PTP expression, and found that atorvastatin, a HMG-CoA reductase inhibitor widely used in the clinic to treat hypercholesterolemia-related cardiovascular diseases, strongly promoted VE-PTP transcription in ECs through activating the VE-PTP promoter and upregulating the expression of the transcription factor, specificity protein 1 (SP1). Additionally, atorvastatin markedly reduced VE-cadherin-Y731 phosphorylation induced by cigarette smoke extract and significantly enhanced stability of endothelial adherens junctions. Together, our findings reveal that atorvastatin up-regulates VE-PTP expression, increases VE-cadherin protein levels, and decreases VE-cadherin-Y731 phosphorylation to strengthen EC adherens junctions and maintain vascular cell monolayer integrity, offering a new mechanism of atorvastatin against CSE-induced disruption of vascular integrity and relevant cardio-cerebrovascular disease.
Collapse
|
37
|
Wu J, Zhang W, Liu X, Wu L, He G, Li P, Guo X, Chen Z, Huang Q. Apocynin protects endothelial cells from endoplasmic reticulum stress-induced apoptosis via IRE1α engagement. Mol Cell Biochem 2018; 449:257-265. [PMID: 29696609 PMCID: PMC7088535 DOI: 10.1007/s11010-018-3362-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/16/2018] [Indexed: 12/22/2022]
Abstract
Endoplasmic reticulum (ER) stress-induced endothelial cell (EC) apoptosis has been implicated in a variety of human diseases. In addition to being regarded as an NADPH oxidase (NOX) inhibitor, apocynin (APO) exhibits an anti-apoptotic effect in various cells. The present study aimed to identify the protective role of apocynin in ER stress-mediated EC apoptosis and the underlying mechanisms. We found that ER stress resulted in a significant increase in c-Jun N-terminal kinase phosphorylation, and elicited caspase 3 cleavage and apoptosis. However, apocynin obviously attenuated EC apoptosis and this effect was partly dependent on ER stress sensor inositol-requiring enzyme 1α (IRE1α). Importantly, apocynin upregulated IRE1α expression in both protein and mRNA levels and promoted the pro-survival XBP1 splicing. Our results suggest that apocynin protects ECs against ER stress-induced apoptosis via IRE1α involvement. These findings may provide a novel mechanistic explanation for the anti-apoptotic effect of apocynin in ER stress.
Collapse
Affiliation(s)
- Jie Wu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijin Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohui Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lili Wu
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Guangzhou, 510515, Guangdong, China
| | - Guangting He
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Peixin Li
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaohua Guo
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Guangzhou, 510515, Guangdong, China
| | - Zhongqing Chen
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Basic Medical Sciences, Southern Medical University, No. 1023, South Shatai Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
38
|
Zhou H, Jin C, Cui L, Xing H, Liu J, Liao W, Liao H, Yu Y. HMGB1 contributes to the irradiation-induced endothelial barrier injury through receptor for advanced glycation endproducts (RAGE). J Cell Physiol 2018; 233:6714-6721. [PMID: 29215715 DOI: 10.1002/jcp.26341] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
This study aimed to investigate whether HMGB1 (high mobility group box-1 protein) and receptor for advanced glycation end products (RAGE) were involved in the irradiation-induced endothelial barrier damage and their mechanism. We constructed the damage model of endothelium barrier model with bEnd.3 cells. The permeability of endothelial barrier was detected by sodium fluorescein (Na-F) permeation test, and the irradiation dose which could induce permeability transition was determined by being exposed to different irradiation doses (5, 10, 15, 20 Gy). MTT assay was applied to detect cell viability under different concentrations of HMGB1, glycyrrhizic acid (GA, a specific inhibitor of HMGB1), and FPS-ZM1 (a blood-brain-barrier permeant blocker of RAGE V domain-mediated ligand binding). The expression of HMGB1, RAGE, and related molecules involved in MAPK signaling pathway, MMP-2, MMP-9, ZO-1, and claudin 5 of differently treated groups were measured by qRT-PCR, western blot, and immunofluorescence. Cells possessed stable endothelial barrier function on 4-7 days after seeded on transwell plates. The permeability of endothelial barrier would change under at least 10 Gy radiation. Both radiation and HMGB1 treatment alone could improve the permeability. After irradiation, the expressions of HMGB1 and RAGE increased and MAPK signal pathway was activated. Meanwhile, MMP-2 and MMP-9 were overexpressed, while the expression of tight junction proteins ZO-1 and claudin 5 was decreased. Radiation could activate MAPK signaling pathway through promoting the expression of HMGB1 and RAGE, which further led to endothelial barrier injury and changed its permeability.
Collapse
Affiliation(s)
- Haihong Zhou
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Congli Jin
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lili Cui
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huaijie Xing
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jun Liu
- Department of Neurology, SUN YAT-SEN Medical Hospital, SUN YAT-SEN University, Guangzhou, Guangdong, China
| | - Wang Liao
- Department of Neurology, SUN YAT-SEN Medical Hospital, SUN YAT-SEN University, Guangzhou, Guangdong, China
| | - Haojie Liao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yangsheng Yu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
39
|
Sirt1 Protects Endothelial Cells against LPS-Induced Barrier Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4082102. [PMID: 29209448 PMCID: PMC5676476 DOI: 10.1155/2017/4082102] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/16/2017] [Accepted: 09/12/2017] [Indexed: 01/10/2023]
Abstract
Sepsis is a threatening health problem and characterized by microvascular dysfunction. In this study, we verified that LPS caused the downregulation of Sirt1 and the hyperpermeability of endothelial cells. Inhibition of Sirt1 with ex527 or Sirt1 siRNA displayed a higher permeability, while activation of Sirt1 with SRT1720 reversed the LPS-induced hyperpermeability, formation of fiber stress, and disruption of VE-cadherin distribution. In pulmonary microvascular vein endothelial cells isolated from wild-type mice, Sirt1 was attenuated upon LPS, while Sirt1 was preserved in a receptor of advanced glycation end product-knockout mice. The RAGE antibody could also diminish the downregulation and ubiquitination of Sirt1 in LPS-exposed human umbilical vein endothelial cells. An LPS-induced decrease in Sirt1 activity was attenuated by the RAGE antibody and TLR4 inhibitor. In vivo study also demonstrated the attenuating role of Sirt1 and RAGE knockout in LPS-induced increases in dextran leakage of mesenteric venules. Furthermore, activation of Sirt1 prevented LPS-induced decreases in the activity and expression of superoxide dismutase 2, as well as the increases in NADPH oxidase 4 and reactive oxygen species, while inhibition of Sirt1 aggravated the SOD2 decline. It also demonstrated that Sirt1-deacetylated p53 is required for p53 inactivation, which reversed the downregulation of β-catenin caused by LPS.
Collapse
|