1
|
Wang F, Wang J, Liang X, Wu Z, Xue J, Yin L, Wei L, Zhang X. Ghrelin inhibits myocardial pyroptosis in diabetic cardiomyopathy by regulating ERS and NLRP3 inflammasome crosstalk through the PI3K/AKT pathway. J Drug Target 2024; 32:148-158. [PMID: 38088811 DOI: 10.1080/1061186x.2023.2295268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023]
Abstract
AIMS Endoplasmic reticulum stress(ERS) can induce inflammation mediated by NLRP3 inflammatory bodies and link inflammation with oxidative stress in myocardial tissue. Ghrelin is an endogenous growth hormone-releasing peptide that has been proven to have multiple effects, such as regulating energy metabolism and inhibiting inflammation. However, the role of ghrelin in myocardial injury in diabetic rats and the mechanism have not been reported. RESULTS We found that ghrelin could improve endoplasmic reticulum stress and inflammatory pyroptosis in the myocardial tissue of diabetic rats and reduce ERS and NLRP3 inflammasome crosstalk in H9C2 cardiomyocytes. Interestingly, ghrelin could activate the PI3K/AKT signalling pathway, playing a role in inhibiting endoplasmic reticulum stress and reducing the expression of pyroptosis-related proteins. However, these protective effects could be largely eliminated by LY294002. CONCLUSIONS In summary, we demonstrated that ghrelin inhibited myocardial pyroptosis in diabetic cardiomyopathy by regulating ERS and NLRP3 inflammasome crosstalk through the PI3K/AKT pathway. Our results provide new insights into the mechanism of diabetic myocardial injury induced by high glucose and high palmitic acid and ghrelin-mediated anti-inflammatory protection and provide potential therapeutic targets and strategies for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Fan Wang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingzhi Wang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinfang Liang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zixuan Wu
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxin Xue
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lingyu Yin
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lai Wei
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaohui Zhang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
2
|
Li S, Zhou Y, Li K, Liu L, Fang M, Gao H. Inhibition of circDGKZ ameliorates myocardial ischemia/reperfusion injury by targeting miR-345-5p/TLR4. ESC Heart Fail 2024; 11:2730-2741. [PMID: 38725137 PMCID: PMC11424286 DOI: 10.1002/ehf2.14809] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/25/2024] [Accepted: 04/01/2024] [Indexed: 09/27/2024] Open
Abstract
AIMS This study aims to explore the molecular mechanism of circular RNAs' (circRNAs) potential involvement in myocardial ischaemia-reperfusion injury (MIRI). METHODS AND RESULTS Differently expressed genes in myocardial infarction (MI) were identified by screening the GEO database. Serum was collected from MI patients and healthy volunteers (n = 5 for each group). AC16 cells were cultured and exposed to hypoxia/reperfusion (H/R) treatment for the cell experiments. Then candidate genes were validated in human serum and the H/R model. Quantitative real-time PCR and western blot were used to detect expression of key molecules such as circDGKZ, miR-345-5p, and Toll-like receptor 4 (TLR4), as well as pyroptosis markers such as NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), ASC, C-caspase1, interleukin (IL)-1β, and IL-18. CircDGKZ was positively correlated in human serum (P < 0.05) and in AC16 cells (P < 0.01). Knockdown of circDGKZ inhibited cardiomyocyte pyroptosis and the TLR4/nuclear factor kappa B (NF-κB) signalling pathway (all P < 0.05). A luciferase assay was used to detect the molecule interaction. MiR-345-5p was regulated by circDGKZ and regulated TLR4 in cardiomyocytes both through direct interaction (P < 0.01). The stability and distribution of circRNA or linear RNA were examined by subcellular localization and RNA decay assays. CircDGKZ was stably expressed in cardiomyocytes and mainly distributed in the cytoplasm (P < 0.01). Knockdown of circDGKZ also promoted the degradation of NLRP3 by inducing autophagy (P < 0.05). MIRI rat models were constructed (n = 5 for each group), and the cellular results were further confirmed in rat models (P < 0.05). CONCLUSIONS Knockdown of circDGKZ interrupted pyroptosis and induced autophagy of cardiomyocytes via regulating miR-345-5p/TLR4/NF-κB.
Collapse
Affiliation(s)
- Shiliang Li
- Division of Cardiothoracic and Vascular SurgeryTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Yan Zhou
- Department of OtolaryngologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - KunSheng Li
- Department of Cardiovascular SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu ProvinceChina
| | - Lu Liu
- Department of PharmacyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Ming Fang
- Health Management CenterUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanHubei ProvinceChina
| | - Hongfeng Gao
- Department of Emergency MedicineWuhan Wuchang Hospital, Wuchang Hospital Affiliated to Wuhan University of Science and TechnologyNo. 116, Yangyuan StreetWuhan430000Hubei ProvinceChina
| |
Collapse
|
3
|
Sullivan R, Hou J, Yu L, Wilk B, Sykes J, Biernaski H, Butler J, Kovacs M, Hicks J, Thiessen JD, Dharmakumar R, Prato FS, Wisenberg G, Luyt LG, Dhanvantari S. Design, Synthesis, and Preclinical Evaluation of a High-Affinity 18F-Labeled Radioligand for Myocardial Growth Hormone Secretagogue Receptor Before and After Myocardial Infarction. J Nucl Med 2024; 65:1633-1639. [PMID: 39266294 DOI: 10.2967/jnumed.124.267578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024] Open
Abstract
The peptide hormone ghrelin is produced in cardiomyocytes and acts through the myocardial growth hormone secretagogue receptor (GHSR) to promote cardiomyocyte survival. Administration of ghrelin may have therapeutic effects on post-myocardial infarction (MI) outcomes. Therefore, there is a need to develop molecular imaging probes that can track the dynamics of GHSR in health and disease to better predict the effectiveness of ghrelin-based therapeutics. We designed a high-affinity GHSR ligand labeled with 18F for imaging by PET and characterized its in vivo properties in a canine model of MI. Methods: We rationally designed and radiolabeled with 18F a quinazolinone derivative ([18F]LCE470) with subnanomolar binding affinity to GHSR. We determined the sensitivity and in vivo and ex vivo specificity of [18F]LCE470 in a canine model of surgically induced MI using PET/MRI, which allowed for anatomic localization of tracer uptake and simultaneous determination of global cardiac function. Uptake of [18F]LCE470 was determined by time-activity curve and SUV analysis in 3 regions of the left ventricle-area of infarct, territory served by the left circumflex coronary artery, and remote myocardium-over a period of 1.5 y. Changes in cardiac perfusion were tracked by [13N]NH3 PET. Results: The receptor binding affinity of LCE470 was measured at 0.33 nM, the highest known receptor binding affinity for a radiolabeled GHSR ligand. In vivo blocking studies in healthy hounds and ex vivo blocking studies in myocardial tissue showed the specificity of [18F]LCE470, and sensitivity was demonstrated by a positive correlation between tracer uptake and GHSR abundance. Post-MI changes in [18F]LCE470 uptake occurred independently of perfusion tracer distributions and changes in global cardiac function. We found that the regional distribution of [18F]LCE470 within the left ventricle diverged significantly within 1 d after MI and remained that way throughout the 1.5-y duration of the study. Conclusion: [18F]LCE470 is a high-affinity PET tracer that can detect changes in the regional distribution of myocardial GHSR after MI. In vivo PET molecular imaging of the global dynamics of GHSR may lead to improved GHSR-based therapeutics in the treatment of post-MI remodeling.
Collapse
Affiliation(s)
- Rebecca Sullivan
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Jinqiang Hou
- Lakehead University and Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| | - Lihai Yu
- London Regional Cancer Program, London, Ontario, Canada
| | - Benjamin Wilk
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jane Sykes
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Heather Biernaski
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - John Butler
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Michael Kovacs
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Justin Hicks
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jonathan D Thiessen
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | | | - Frank S Prato
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Gerald Wisenberg
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Leonard G Luyt
- London Regional Cancer Program, London, Ontario, Canada
- Departments of Chemistry, Oncology, and Medical Imaging, Western University, London, Ontario, Canada
| | - Savita Dhanvantari
- Imaging Research Program, Lawson Health Research Institute, London, Ontario, Canada;
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| |
Collapse
|
4
|
Liao SS, Zhang LL, Zhang YG, Luo J, Kadier T, Ding K, Chen R, Meng QT. Ghrelin alleviates intestinal ischemia-reperfusion injury by activating the GHSR-1α/Sirt1/FOXO1 pathway. FASEB J 2024; 38:e23681. [PMID: 38814725 DOI: 10.1096/fj.202302155rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 06/01/2024]
Abstract
Ischemia-reperfusion (IR) injury is primarily characterized by the restoration of blood flow perfusion and oxygen supply to ischemic tissue and organs, but it paradoxically leads to tissue injury aggravation. IR injury is a challenging pathophysiological process that is difficult to avoid clinically and frequently occurs during organ transplantation, surgery, shock resuscitation, and other processes. The major causes of IR injury include increased levels of free radicals, calcium overload, oxidative stress, and excessive inflammatory response. Ghrelin is a newly discovered brain-intestinal peptide with anti-inflammatory and antiapoptotic effects that improve blood supply. The role and mechanism of ghrelin in intestinal ischemia-reperfusion (IIR) injury remain unclear. We hypothesized that ghrelin could attenuate IIR-induced oxidative stress and apoptosis. To investigate this, we established IIR by using a non-invasive arterial clip to clamp the root of the superior mesenteric artery (SMA) in mice. Ghrelin was injected intraperitoneally at a dose of 50 μg/kg 20 min before IIR surgery, and [D-Lys3]-GHRP-6 was injected intraperitoneally at a dose of 12 nmol/kg 20 min before ghrelin injection. We mimicked the IIR process with hypoxia-reoxygenation (HR) in Caco-2 cells, which are similar to intestinal epithelial cells in structure and biochemistry. Our results showed that ghrelin inhibited IIR/HR-induced oxidative stress and apoptosis by activating GHSR-1α. Moreover, it was found that ghrelin activated the GHSR-1α/Sirt1/FOXO1 signaling pathway. We further inhibited Sirt1 and found that Sirt1 was critical for ghrelin-mediated mitigation of IIR/HR injury. Overall, our data suggest that pretreatment with ghrelin reduces oxidative stress and apoptosis to attenuate IIR/HR injury by binding with GHSR-1α to further activate Sirt1.
Collapse
Affiliation(s)
- Shi-Shi Liao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Le-le Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Guo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Luo
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tulanisa Kadier
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Katayama H, Kaiya H. Identification, chemical synthesis, and receptor binding of a reptilian gecko ghrelin. J Pept Sci 2024; 30:e3567. [PMID: 38268104 DOI: 10.1002/psc.3567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/26/2024]
Abstract
Ghrelin is known to be a gastrointestinal peptide hormone in vertebrates. It has a unique posttransrational modification, octanoylation, at the Ser side chain of the third position. In this study, we identified the genes encoding ghrelin and its receptor from the Schlegel's Japanese gecko Gekko japonicus. The C-terminal residue of gecko ghrelin was His, although the chemical synthesis method for the O-octanoyl peptide with a C-terminal His residue has not yet been well-established. Acyl-ghrelin has been synthesized using a Ser derivative without side chain protecting group in the solid-phase peptide synthesis, although this synthetic strategy has not yet been well-established. Here we show the efficient synthetic method with minimal side reactions, and G. japonicus ghrelin could be obtained in good yield. This would be useful and applicable to the synthesis of ghrelin from other animal species. The gecko ghrelin receptor was expressed in HEK 293 cells, which was fully responsive to the synthetic gecko ghrelin. These results indicate that the ghrelin system similar to mammals also exists in a reptilian gecko, G. japonicus.
Collapse
Affiliation(s)
- Hidekazu Katayama
- Department of Bioengineering, School of Engineering, Tokai University, Hiratsuka, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
- Research Division of Drug Discovery, Grandsoul Research Institute for Immunology Inc., Nara, Japan
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| |
Collapse
|
6
|
Majid Z, Muhammad-Baqir B, Al-Shimerty DF, Hadi NR. The possible cardioprotective effect of ghrelin during experimental endotoxemia in mice. J Med Life 2024; 17:486-491. [PMID: 39144689 PMCID: PMC11320619 DOI: 10.25122/jml-2023-0228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/04/2023] [Indexed: 08/16/2024] Open
Abstract
This study aimed to evaluate the cardioprotective effects of ghrelin in septic mice, focusing on its anti-inflammatory and antioxidant properties. Thirty-five male Swiss mice (8-12 weeks old, 23-33g) were randomly assigned to five groups (n = 7 each): (1) Normal, fed usual diets, (2) Sham, subjected to anesthesia and laparotomy, (3) Sepsis, subjected to cecal ligation and puncture, (4) Vehicle, given an equivalent volume of intraperitoneal saline injections immediately after cecal ligation and puncture, and (5) Ghrelin-treated, administered 80 µg/kg ghrelin intraperitoneal injections immediately following cecal ligation and puncture. Serum levels of tumor necrosis factor-alpha (TNF-α), macrophage migration inhibitory factor (MIF), toll-like receptor 4 (TLR4), and 8-epi-prostaglandin F2 alpha (8-epi-PGF2α) were measured. The extent of cardiac damage was also evaluated histologically. The mean serum levels of TNF-α, MIF, TLR4, and 8-epi-PGF2α levels were significantly higher in the sepsis and vehicle groups than in the normal and sham groups. The levels were significantly lower in the ghrelin-treated group than in the vehicle and sepsis groups. Histological analysis revealed normal myocardial architecture in the normal and sham groups, whereas the sepsis and vehicle groups had severe myocardial injury. The ghrelin-treated group displayed histological features similar to the sham group, indicating reduced myocardial damage. Ghrelin ameliorated sepsis-induced cardiotoxicity in mice by exhibiting strong anti-inflammatory and antioxidant effects. These findings suggest that ghrelin may be a promising therapeutic candidate for the prevention of sepsis-induced cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | - Najah Rayish Hadi
- Department of Pharmacology & Therapeutics, Faculty of Medicine, University of Kufa, Kufa, Iraq
| |
Collapse
|
7
|
Zhang L, Zhao S, Wang Y. Diannexin alleviates myocardial ischemia-reperfusion injury by orchestrating cardiomyocyte oxidative damage, macrophage polarization and fibrotic process by TLR4-NF-kB-mediated inactivation of NLRP3 inflammasome. Int Immunopharmacol 2024; 130:111668. [PMID: 38417368 DOI: 10.1016/j.intimp.2024.111668] [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: 11/12/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 03/01/2024]
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a pathogenic mechanism of myocardial infarction and heart failure, constituting a major health concern globally. Diannexin is a homodimer of recombinant human annexin V and elicits important roles in several I/R injuries. Nevertheless, its function in MI/R remains elusive. Here, Diannexin alleviated simulated I/R (SI/R)-induced cardiomyocyte death and oxidative injury by increasing cell viability and inhibiting cell apoptosis, ROS, lactate dehydrogenase, malondialdehyde production and anti-oxidative SOD activity. Diannexin inhibited SI/R-induced expression of fibrotic protein collagen I and collagen III. Furthermore, Diannexin suppressed LPS-induced macrophage polarization towards pro-inflammatory M1-like phenotype and enhanced IL-4-evoked anti-inflammatory M2 polarization. Concomitantly, Diannexin inhibited SI/R exposure-induced macrophage polarization to M1 subtypes. Importantly, conditioned medium (CM) from SI/R-stimulated macrophages evoked cardiomyocyte apoptosis, which was reversed when cells were co-cultured with CM from Diannexin-treated macrophages under SI/R conditions. Mechanically, the activation of TLR4/NF-κB/NLRP3 inflammasome signaling in SI/R-treated cells was mitigated by Diannexin. Reactivating this pathway antagonized the protective effects of Diannexin on SI/R-induced cardiomyocyte oxidative injury, fibrotic protein expression and macrophage polarization and M1 macrophage-induced apoptosis of cardiomyocytes. In vivo, Diannexin alleviated abnormal cardiac structure, dysfunction and collagen position in MI/R mice. Additionally, Diannexin reduced M1-polarized and elevated M2-polarized macrophages in heart tissues at five days post-MI/R. The activation of TLR4/NF-κB/NLRP3 inflammasome pathway in MI/R mice was attenuated after Diannexin administration. Together, Diannexin may alleviate the development of MI/R injury by directly regulating cardiomyocyte oxidative injury, fibrotic potential and indirectly affecting macrophage polarization-mediated cardiomyocyte apoptosis, indicating a promising therapeutic strategy for MI/R.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Songlin Zhao
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaqi Wang
- Department of Clinical Laboratory Medicine, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
8
|
Song R, Baker TL, Watters JJ, Kumar S. Obstructive Sleep Apnea-Associated Intermittent Hypoxia-Induced Immune Responses in Males, Pregnancies, and Offspring. Int J Mol Sci 2024; 25:1852. [PMID: 38339130 PMCID: PMC10856042 DOI: 10.3390/ijms25031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/22/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Obstructive sleep apnea (OSA), a respiratory sleep disorder associated with cardiovascular diseases, is more prevalent in men. However, OSA occurrence in pregnant women rises to a level comparable to men during late gestation, creating persistent effects on both maternal and offspring health. The exact mechanisms behind OSA-induced cardiovascular diseases remain unclear, but inflammation and oxidative stress play a key role. Animal models using intermittent hypoxia (IH), a hallmark of OSA, reveal several pro-inflammatory signaling pathways at play in males, such as TLR4/MyD88/NF-κB/MAPK, miRNA/NLRP3, and COX signaling, along with shifts in immune cell populations and function. Limited evidence suggests similarities in pregnancies and offspring. In addition, suppressing these inflammatory molecules ameliorates IH-induced inflammation and tissue injury, providing new potential targets to treat OSA-associated cardiovascular diseases. This review will focus on the inflammatory mechanisms linking IH to cardiovascular dysfunction in males, pregnancies, and their offspring. The goal is to inspire further investigations into the understudied populations of pregnant females and their offspring, which ultimately uncover underlying mechanisms and therapeutic interventions for OSA-associated diseases.
Collapse
Affiliation(s)
- Ruolin Song
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Tracy L. Baker
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Jyoti J. Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
| | - Sathish Kumar
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA; (R.S.); (T.L.B.); (J.J.W.)
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53792, USA
| |
Collapse
|
9
|
Zhang X, Zeng Z, Liu Y, Liu D. Emerging Relevance of Ghrelin in Programmed Cell Death and Its Application in Diseases. Int J Mol Sci 2023; 24:17254. [PMID: 38139082 PMCID: PMC10743592 DOI: 10.3390/ijms242417254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Ghrelin, comprising 28 amino acids, was initially discovered as a hormone that promotes growth hormones. The original focus was on the effects of ghrelin on controlling hunger and satiation. As the research further develops, the research scope of ghrelin has expanded to a wide range of systems and diseases. Nevertheless, the specific mechanisms remain incompletely understood. In recent years, substantial studies have demonstrated that ghrelin has anti-inflammatory, antioxidant, antiapoptotic, and other effects, which could affect the signaling pathways of various kinds of programmed cell death (PCD) in treating diseases. However, the regulatory mechanisms underlying the function of ghrelin in different kinds of PCD have not been thoroughly illuminated. This review describes the relationship between ghrelin and four kinds of PCD (apoptosis, necroptosis, autophagy, and pyroptosis) and then introduces the clinical applications based on the different features of ghrelin.
Collapse
Affiliation(s)
- Xue Zhang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Zihan Zeng
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Yaning Liu
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Dan Liu
- School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
10
|
Ramalingam V. NLRP3 inhibitors: Unleashing their therapeutic potential against inflammatory diseases. Biochem Pharmacol 2023; 218:115915. [PMID: 37949323 DOI: 10.1016/j.bcp.2023.115915] [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: 07/23/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome has been linked to the release of pro-inflammatory cytokines and is essential for innate defence against infection and danger signals. These secreted cytokines improve the inflammatory response caused by tissue damage and associated inflammation. Consequently, the development of NLRP3 inflammasome inhibitors are viable option for the treatment of diverse inflammatory disorders. The significant anti-inflammatory effects of the NLRP3 inhibitors have severe side effects. Hence, the application of NLRP3 inhibitors against inflammatory disease has not yet been understood and most of the developed inhibitors are unsuccessful in clinical trials. The processes behind the NLRP3 complex, priming, and activation are the main emphasis of this review, which also covers therapeutical inhibitors of the NLRP3 inflammasome and potential therapeutic strategies for directing the NLRP3 inflammasome towards clinical development.
Collapse
Affiliation(s)
- Vaikundamoorthy Ramalingam
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
11
|
Wang Z, Zhu D, Zhang Y, Xia F, Zhu J, Dai J, Zhuge X. Extracellular vesicles produced by avian pathogenic Escherichia coli (APEC) activate macrophage proinflammatory response and neutrophil extracellular trap (NET) formation through TLR4 signaling. Microb Cell Fact 2023; 22:177. [PMID: 37689682 PMCID: PMC10492386 DOI: 10.1186/s12934-023-02171-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/08/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Avian pathogenic Escherichia coli (APEC) is the major pathogen causing important avian diseases in poultry. As an important subtype of extraintestinal pathogenic E. coli, APEC has zoonotic potential and is considered a foodborne pathogen. APEC extracellular vesicles (EVs) may play vital roles in the interaction of the pathogen with its host cells. However, the precise roles played by APEC EVs are still not completely clear, especially in immune cells. RESULTS In this study, we investigated the relationships between APEC EVs and immune cells. The production and characteristics of the EVs of APEC isolate CT265 were identified. Toll like receptor 4 (TLR4) triggered the cellular immune responses when it interacted with APEC EVs. APEC EVs induced a significant release of proinflammatory cytokines in THP-1 macrophages. APEC EVs induced the macrophage inflammatory response via the TLR4/MYD88/NF-κB signaling pathway, which participated in the activation of the APEC-EV-induced NLRP3 inflammasome. However, the loss of lipopolysaccharide (LPS) from APEC EVs reduced the activation of the NLRP3 inflammasome mediated by TLR4/MYD88/NF-κB signaling. Because APEC EVs activated the macrophage inflammatory response and cytokines release, we speculated that the interaction between APEC EVs and macrophages activated and promoted neutrophil migration during APEC extraintestinal infection. This study is the first to report that APEC EVs induce the formation of neutrophil extracellular traps (NETs) and chicken heterophil extracellular traps. Treatment with APEC EVs induced SAPK/JNK activation in neutrophils. The inhibition of TLR4 signaling suppressed APEC-EV-induced NET formation. However, although APEC EVs activated the immune response of macrophages and initiated NET formation, they also damaged macrophages, causing their apoptosis. The loss of LPS from APEC EVs did not prevent this process. CONCLUSION APEC-derived EVs induced inflammatory responses in macrophages and NETs in neutrophils, and that TLR4 was involved in the APEC-EV-activated inflammatory response. These findings provided a basis for the further study of APEC pathogenesis.
Collapse
Affiliation(s)
- Zhongxing Wang
- Key Lab of Animal Bacteriology, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Ministry of Agriculture, Nanjing Agricultural University, No.1 Weigang road, Nanjing, 210095, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, No.9 Seyuan road, Nantong, Jiangsu, 226019, P.R. China
| | - Dongyu Zhu
- Key Lab of Animal Bacteriology, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Ministry of Agriculture, Nanjing Agricultural University, No.1 Weigang road, Nanjing, 210095, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, No.9 Seyuan road, Nantong, Jiangsu, 226019, P.R. China
| | - Yuting Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, No.9 Seyuan road, Nantong, Jiangsu, 226019, P.R. China
| | - Fufang Xia
- Key Lab of Animal Bacteriology, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Ministry of Agriculture, Nanjing Agricultural University, No.1 Weigang road, Nanjing, 210095, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, No.9 Seyuan road, Nantong, Jiangsu, 226019, P.R. China
| | - Jiaying Zhu
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jianjun Dai
- Key Lab of Animal Bacteriology, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Ministry of Agriculture, Nanjing Agricultural University, No.1 Weigang road, Nanjing, 210095, China.
- College of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiangkai Zhuge
- Key Lab of Animal Bacteriology, MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Ministry of Agriculture, Nanjing Agricultural University, No.1 Weigang road, Nanjing, 210095, China.
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, No.9 Seyuan road, Nantong, Jiangsu, 226019, P.R. China.
| |
Collapse
|
12
|
Ibrahim HA, Abd El-Alim AEAF, El-Hafeez MA, Metwally MMM, Khamis T, Galal AAA. Baicalein prevents capecitabine-induced heart damage in female Wistar rats and enhances its anticancer potential in MCF-7 breast cancer cells. Life Sci 2023; 319:121523. [PMID: 36842762 DOI: 10.1016/j.lfs.2023.121523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
AIM We investigated the ability of baicalein (BAI) to enhance the anticancer potential of capecitabine (CAP) in the MCF-7 cell line and its protective effect on CAP-induced cardiotoxicity in female Wistar rats. METHODS AND KEY FINDINGS In vitro study involved evaluating the effect of BAI and/or CAP on cell viability, cell cycle progression, and BAX and Bcl2 gene expression in MCF-7 cells. Co-treatment of BAI with CAP significantly reduced the viability of MCF-7 cells, improved their cytotoxic effect, markedly elevated the percentage of the sub-G1 population, drastically reduced the G2/M population, and significantly altered the mRNA expression of BAX and Bcl2 genes compared with each treatment alone. In vivo study revealed that the oral administration of CAP (140 mg/kg BW) to adult female rats significantly elevated the levels of serum creatine kinase-myocardial band (CK-MB), lactate dehydrogenase (LDH), tumor necrosis factor (TNF)-α, and interleukin (IL)-1β and cardiac TNF-α, IL-1β malondialdehyde (MDA) concentration, whereas it reduced the serum and cardiac total antioxidant capacity (TAC), level of cardiac glutathione (GSH) and activity of glutathione peroxidase (GPx) with a vast array of circulatory, inflammatory, degenerative, and necrotic alterations in the cardiac tissue. Furthermore, CAP administration significantly upregulated the mRNA expression of NF-κB, TLR4, MyD88, ATF6, CHOP, and JNK genes. Concurrent administration of BAI (200 mg/kg BW) and CAP significantly improved the biochemical alterations and cardiac oxidant/antioxidant status and architecture. In addition, it modulated the TLR4/MyD88/NF-κB pathway and endoplasmic reticulum stress. SIGNIFICANCE Altogether, BAI can augment the anticancer potential of CAP and alleviate its cardiotoxic effects during cancer treatment.
Collapse
Affiliation(s)
- Hosny A Ibrahim
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | | | - Mai Abd El-Hafeez
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed M M Metwally
- Pathology department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Tarek Khamis
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Azza A A Galal
- Pharmacology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt.
| |
Collapse
|
13
|
Liu XY, Wei DG, Li RS. Ghrelin attenuates inflammation in diabetic lung disease by TLR4 pathway in vivo and in vitro. BMJ Open Diabetes Res Care 2023; 11:11/2/e003027. [PMID: 37085277 PMCID: PMC10123865 DOI: 10.1136/bmjdrc-2022-003027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 03/11/2023] [Indexed: 04/23/2023] Open
Abstract
INTRODUCTION Diabetic lung disease is already known as one of the diabetes complications, but report on its therapeutic strategy is rare. The present study aimed to add novel therapeutic strategy for diabetic lung disease, to reveal the protective effect of ghrelin on diabetic lung disease both in vivo and in vitro, and to discuss its probable molecular mechanism. RESEARCH DESIGN AND METHODS Diabetic mice and 16HBE cells were our research objects. We surveyed the effect of ghrelin on streptozotocin-induced lung tissue morphology changes by H&E staining. Furthermore, the changes of proinflammatory cytokines (interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α)) were detected by ELISA. To expound the molecular mechanism, we detected critical proteins of TLR4 pathway and observed their changes by immunohistochemistry (IHC), real-time PCR and western blot analysis in vivo and in vitro, respectively. RESULTS The results of H&E staining showed that pathological alterations of the lung induced by hyperglycemia were ameliorated by ghrelin. The results of ELISA demonstrated that the elevated levels of IL-1β and TNF-α induced by hyperglycemia turned to decrease in the lung after ghrelin treatment. In the results of IHC, real-time PCR and western blot analysis, we found that the TLR4 pathway was elevated by hyperglycemia or high glucose and is remarkably inhibited by the treatment of ghrelin both in vivo and in vitro. CONCLUSIONS Ghrelin could inhibit inflammation of diabetic lung disease by regulating the TLR4 pathway. This study might affect research on diabetic lung disease, and the therapeutic potential of ghrelin for diabetic lung disease is worth considering.
Collapse
Affiliation(s)
- Xiao-Yan Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Dong-Guang Wei
- Department of Pulmonary and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Rong-Shan Li
- Department of Nephrology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
14
|
Inceu AI, Neag MA, Craciun AE, Buzoianu AD. Gut Molecules in Cardiometabolic Diseases: The Mechanisms behind the Story. Int J Mol Sci 2023; 24:3385. [PMID: 36834796 PMCID: PMC9965280 DOI: 10.3390/ijms24043385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the most common cause of morbidity and mortality worldwide. Diabetes mellitus increases cardiovascular risk. Heart failure and atrial fibrillation are associated comorbidities that share the main cardiovascular risk factors. The use of incretin-based therapies promoted the idea that activation of alternative signaling pathways is effective in reducing the risk of atherosclerosis and heart failure. Gut-derived molecules, gut hormones, and gut microbiota metabolites showed both positive and detrimental effects in cardiometabolic disorders. Although inflammation plays a key role in cardiometabolic disorders, additional intracellular signaling pathways are involved and could explain the observed effects. Revealing the involved molecular mechanisms could provide novel therapeutic strategies and a better understanding of the relationship between the gut, metabolic syndrome, and cardiovascular diseases.
Collapse
Affiliation(s)
- Andreea-Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Maria-Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca-Elena Craciun
- Department of Diabetes, and Nutrition Diseases, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Anca-Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| |
Collapse
|
15
|
Russo C, Valle MS, Russo A, Malaguarnera L. The Interplay between Ghrelin and Microglia in Neuroinflammation: Implications for Obesity and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms232113432. [PMID: 36362220 PMCID: PMC9654207 DOI: 10.3390/ijms232113432] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Numerous studies have shown that microglia are capable of producing a wide range of chemokines to promote inflammatory processes within the central nervous system (CNS). These cells share many phenotypical and functional characteristics with macrophages, suggesting that microglia participate in innate immune responses in the brain. Neuroinflammation induces neurometabolic alterations and increases in energy consumption. Microglia may constitute an important therapeutic target in neuroinflammation. Recent research has attempted to clarify the role of Ghre signaling in microglia on the regulation of energy balance, obesity, neuroinflammation and the occurrence of neurodegenerative diseases. These studies strongly suggest that Ghre modulates microglia activity and thus affects the pathophysiology of neurodegenerative diseases. This review aims to summarize what is known from the current literature on the way in which Ghre modulates microglial activity during neuroinflammation and their impact on neurometabolic alterations in neurodegenerative diseases. Understanding the role of Ghre in microglial activation/inhibition regulation could provide promising strategies for downregulating neuroinflammation and consequently for diminishing negative neurological outcomes.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
| | - Maria Stella Valle
- Laboratory of Neuro-Biomechanics, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Antonella Russo
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy
- Correspondence:
| |
Collapse
|
16
|
Molecular Mechanisms and Health Benefits of Ghrelin: A Narrative Review. Nutrients 2022; 14:nu14194191. [PMID: 36235843 PMCID: PMC9572668 DOI: 10.3390/nu14194191] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/26/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022] Open
Abstract
Ghrelin, an endogenous brain-gut peptide, is secreted in large quantities, mainly from the stomach, in humans and rodents. It can perform the biological function of activating the growth hormone secretagogue receptor (GHSR). Since its discovery in 1999, ample research has focused on promoting its effects on the human appetite and pleasure-reward eating. Extensive, in-depth studies have shown that ghrelin is widely secreted and distributed in tissues. Its role in neurohumoral regulation, such as metabolic homeostasis, inflammation, cardiovascular regulation, anxiety and depression, and advanced cancer cachexia, has attracted increasing attention. However, the effects and regulatory mechanisms of ghrelin on obesity, gastrointestinal (GI) inflammation, cardiovascular disease, stress regulation, cachexia treatment, and the prognosis of advanced cancer have not been fully summarized. This review summarizes ghrelin's numerous effects in participating in a variety of biochemical pathways and the clinical significance of ghrelin in the regulation of the homeostasis of organisms. In addition, potential mechanisms are also introduced.
Collapse
|
17
|
Zhang H, Huang C, Zhang D, Zhu Y. Pioglitazone Protects Against Hypoxia-Induced Cardiomyocyte Apoptosis Through Inhibiting NLRP3/Caspase-1 Pathway in vivo and in vitro. Int Heart J 2022; 63:893-903. [PMID: 36104240 DOI: 10.1536/ihj.21-404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study aims to explore the underlying mechanisms of how Pioglitazone (Pio) affects myocardial ischemia-reperfusion (I/R) injury. In this study, after pretreatment of Pio, the pathologic change of myocardial tissues was measured via hematoxylin and eosin staining. The release of lactate dehydrogenase (LDH), superoxide dismutase (SOD), and nitric oxide (NO) were measured. The cardiomyocyte apoptosis was detected via TUNEL assay and flow cytometry assay. The mitochondrial membrane potential (ΔΨm) was estimated using the JC-1 probe. The release of cytochrome c in mitochondria and the translocation of cytochrome c in the cytosol were measured using western blot. Additionally, apoptosis-associated molecules and NOD-like receptor pyrin domain containing-3 (NLRP3)/caspase-1 pathway-related molecules were measured using western blot, quantitative real-time-polymerase chain reaction, and immunofluorescence staining. Results showed that the pretreatment of Pio significantly decreased myocardial tissue damage. Pio pretreatment inhibited the release of creatine kinase and LDH but promoted NO release in serum and H9c2 cell supernatants. Moreover, the pretreatment of Pio notably alleviated cardiomyocyte apoptosis. Pio pretreatment also maintained the mitochondrial membrane potential and prevented cytochrome c release in H/R-induced cardiomyocytes. Additionally, we confirmed that Pio pretreatment inhibited cardiomyocyte apoptosis via repressing the NLRP3/caspase-1 pathway. In conclusion, our study demonstrated that Pio could inhibit myocardial I/R injury and cardiomyocyte apoptosis by inhibiting the activation of the NLRP3/caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Cardiovascular Medicine, The First Hospital of Yulin
| | - Caihong Huang
- Department of Pediatrics, The First Hospital of Yulin
| | - Dong Zhang
- Department of Cardiovascular Medicine, The First Hospital of Yulin
| | - Yan Zhu
- Department of Cardiovascular Medicine, The First Hospital of Yulin
| |
Collapse
|
18
|
Li HL, Tao-Li, Chen ZQ, Li L. Tanshinone IIA reduces pyroptosis in rats with coronary microembolization by inhibiting the TLR4/MyD88/NF-κB/NLRP3 pathway. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:335-345. [PMID: 36039734 PMCID: PMC9437365 DOI: 10.4196/kjpp.2022.26.5.335] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/14/2022] [Indexed: 11/15/2022]
Abstract
Pyroptosis is an inflammatory form of programmed cell death that is linked with invading intracellular pathogens. Cardiac pyroptosis has a significant role in coronary microembolization (CME), thus causing myocardial injury. Tanshinone IIA (Tan IIA) has powerful cardioprotective effects. Hence, this study aimed to identify the effect of Tan IIA on CME and its underlying mechanism. Forty Sprague–Dawley (SD) rats were randomly grouped into sham, CME, CME + low-dose Tan IIA, and CME + high-dose Tan IIA groups. Except for the sham group, polyethylene microspheres (42 µm) were injected to establish the CME model. The Tan-L and Tan-H groups received intraperitoneal Tan IIA for 7 days before CME. After CME, cardiac function, myocardial histopathology, and serum myocardial injury markers were assessed. The expression of pyroptosis-associated molecules and TLR4/MyD88/NF-κB/NLRP3 cascade was evaluated by qRT-PCR, Western blotting, ELISA, and IHC. Relative to the sham group, CME group's cardiac functions were significantly reduced, with a high level of serum myocardial injury markers, and microinfarct area. Also, the levels of caspase-1 p20, GSDMD-N, IL-18, IL-1β, TLR4, MyD88, p-NF-κB p65, NLRP3, and ASC expression were increased. Relative to the CME group, the Tan-H and Tan-L groups had considerably improved cardiac functions, with a considerably low level of serum myocardial injury markers and microinfarct area. Tan IIA can reduce the levels of pyroptosis-associated mRNA and protein, which may be caused by inhibiting TLR4/MyD88/NF-κB/NLRP3 cascade. In conclusion, Tanshinone IIA can suppress cardiomyocyte pyroptosis probably through modulating the TLR4/MyD88/NF-κB/NLRP3 cascade, lowering cardiac dysfunction, and myocardial damage.
Collapse
Affiliation(s)
- Hao-Liang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascul
| | - Tao-Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascul
| | - Zhi-Qing Chen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascul
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University & Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention & Guangxi Clinical Research Center for Cardio-Cerebrovascul
| |
Collapse
|
19
|
Ma Y, Zhang H, Guo W, Yu L. Potential role of ghrelin in the regulation of inflammation. FASEB J 2022; 36:e22508. [PMID: 35983825 DOI: 10.1096/fj.202200634r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Several diseases are caused or progress due to inflammation. In the past few years, accumulating evidence suggests that ghrelin, a gastric hormone of 28-amino acid residue length, exerts protective effects against inflammation by modulating the related pathways. This review focuses on ghrelin's anti-inflammatory and potential therapeutic effects in neurological, cardiovascular, respiratory, hepatic, gastrointestinal, and kidney disorders. Ghrelin significantly alleviates excessive inflammation and reduces damage to different target organs mainly by reducing the secretion of inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and inhibiting the nuclear factor kappa-B (NF-κB) and NLRP3 inflammasome signaling pathways. Ghrelin also regulates inflammation and apoptosis through the p38 MAPK/c-Jun N-terminal kinase (JNK) signaling pathway; restores cerebral microvascular integrity, and attenuates vascular leakage. Ghrelin activates the phosphoInositide-3 kinase (PI3K)/protein kinase B (Akt) pathway and inhibits inflammatory responses in cardiovascular diseases and acute kidney injury. Some studies show that ghrelin exacerbates colonic and intestinal manifestations of colitis. Interestingly, some inflammatory states, such as non-alcoholic steatohepatitis, inflammatory bowel diseases, and chronic kidney disease, are often associated with high ghrelin levels. Thus, ghrelin may be a potential new therapeutic target for inflammation-related diseases.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haifeng Zhang
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Weiying Guo
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
20
|
Russo C, Morello G, Mannino G, Russo A, Malaguarnera L. Immunoregulation of Ghrelin in neurocognitive sequelae associated with COVID-19: an in silico investigation. Gene 2022; 834:146647. [PMID: 35680023 PMCID: PMC9169425 DOI: 10.1016/j.gene.2022.146647] [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: 02/22/2022] [Revised: 05/16/2022] [Accepted: 06/02/2022] [Indexed: 01/08/2023]
Abstract
Some patients suffering from the new Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) develop an exaggerated inflammatory response triggered by a “cytokine storm” resulting in acute respiratory distress syndrome (ARDS) with the concomitant activation of non-specific inflammatory reactivity in the circulatory system and other organs, leading to multiorgan failure, leaky vasculature, coagulopathies and stroke. Impairment of brain functions may also occur as dysregulations in immune function resulting from neuroendocrine interactions. In this study, we explored, by bioinformatics approaches, the interaction between the multiple inflammatory agents involved in SARS-CoV-2 and Ghrelin (Ghre) together with its receptor GHSR-1A, which are described as anti-inflammatory mediators, in order to investigate what could trigger the hyper-inflammatory response in some SARS-CoV-2 patients. In our analysis, we found several interactions of Ghre and GHSR-1A with SARS-CoV-2 interacting human genes. We observed a correlation between Ghre, angiotensin-converting enzyme 2 ACE2, toll-like receptors 9 (TLR9), and Acidic chitinase (CHIA), whereas its receptor GHSR-1A interacts with chemokine receptor 3 (CXCR3), CCR3, CCR5, CCR7, coagulation factor II (thrombin) receptor-like 1 (F2RL1), vitamin D receptor (VDR), Nucleotide-binding oligomerization domain-containing protein 1 (NOD1) and DDP4 in receptor dipeptidyl peptidase-4. To our knowledge, our findings show, for the first time, that Ghre and GHSR-1A may exert an immunomodulatory function in the course of SARS-Cov-2 infection.
Collapse
Affiliation(s)
- Cristina Russo
- Pathology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giovanna Morello
- Department of Biomedical Science, Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Catania, Italy
| | - Giuliana Mannino
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Physiology section, Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antonella Russo
- Physiology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Lucia Malaguarnera
- Pathology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
21
|
Jiang M, Wan S, Dai X, Ye Y, Hua W, Ma G, Pang X, Wang H, Shi B. Protective effect of ghrelin on intestinal I/R injury in rats. Open Med (Wars) 2022; 17:1308-1317. [PMID: 35937002 PMCID: PMC9307145 DOI: 10.1515/med-2022-0520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
This study aimed to investigate whether ghrelin affected the autophagy and inflammatory response of intestinal intraepithelial lymphocytes (IELs) by regulating the NOD2/Beclin-1 pathway in an intestinal ischemia–reperfusion (I/R) injury model. Twenty hours after implementing the intestinal I/R injury rat model, the small intestine and both lungs were collected for histological analysis. The morphological changes in the intestinal mucosa epithelium and lung tissues were evaluated using hematoxylin-eosin staining. The activity of autophagic vacuoles and organ injury were evaluated using electron microscopy. The cytokine levels (IL-10 and TNF-α) in IEL cells and lung tissue were determined using enzyme-linked immunosorbent assay. RT-qPCR and western blot assays were conducted to check the NOD2, Beclin-1, and ATG16 levels. Ghrelin relieved the I/R-induced destruction of the intestinal mucosa epithelium and lung tissues. Moreover, ghrelin enhanced autophagy in the intestinal epithelium and lungs of I/R rats. In addition, the levels of autophagy-associated proteins (Beclin-1, ATG16, and NOD2) were higher in the ghrelin treatment group than in rats with I/R. Ghrelin reduced significantly the IL-10 and TNF-α levels. However, these changes were reversed by the NOD2 antagonist. In conclusion, ghrelin may relieve I/R-induced acute intestinal mucosal damage, autophagy disorder, and inflammatory response in IELs by regulating the NOD2/Beclin-1 pathway.
Collapse
Affiliation(s)
- Meng Jiang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Shengxia Wan
- Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University , Zhenjiang 212000 , China
| | - Xiaoyong Dai
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Youwen Ye
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Wei Hua
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Guoguang Ma
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University , Shanghai 200032 , China
| | - Xiufeng Pang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Huanhuan Wang
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| | - Bin Shi
- Department of Emergency Intensive Care Unit, Yangpu Hospital, Tongji University , Shanghai 200090 , China
| |
Collapse
|
22
|
Effect of TRAF6 in acute pancreatitis-induced intestinal barrier injury via TLR4/NF-κB signal pathway. Tissue Cell 2022; 76:101792. [DOI: 10.1016/j.tice.2022.101792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/28/2022]
|
23
|
Fernandez Rico C, Konate K, Josse E, Nargeot J, Barrère-Lemaire S, Boisguérin P. Therapeutic Peptides to Treat Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:792885. [PMID: 35252383 PMCID: PMC8891520 DOI: 10.3389/fcvm.2022.792885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) including acute myocardial infarction (AMI) rank first in worldwide mortality and according to the World Health Organization (WHO), they will stay at this rank until 2030. Prompt revascularization of the occluded artery to reperfuse the myocardium is the only recommended treatment (by angioplasty or thrombolysis) to decrease infarct size (IS). However, despite beneficial effects on ischemic lesions, reperfusion leads to ischemia-reperfusion (IR) injury related mainly to apoptosis. Improvement of revascularization techniques and patient care has decreased myocardial infarction (MI) mortality however heart failure (HF) morbidity is increasing, contributing to the cost-intense worldwide HF epidemic. Currently, there is no treatment for reperfusion injury despite promising results in animal models. There is now an obvious need to develop new cardioprotective strategies to decrease morbidity/mortality of CVD, which is increasing due to the aging of the population and the rising prevalence rates of diabetes and obesity. In this review, we will summarize the different therapeutic peptides developed or used focused on the treatment of myocardial IR injury (MIRI). Therapeutic peptides will be presented depending on their interacting mechanisms (apoptosis, necroptosis, and inflammation) reported as playing an important role in reperfusion injury following myocardial ischemia. The search and development of therapeutic peptides have become very active, with increasing numbers of candidates entering clinical trials. Their optimization and their potential application in the treatment of patients with AMI will be discussed.
Collapse
Affiliation(s)
- Carlota Fernandez Rico
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Karidia Konate
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emilie Josse
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Prisca Boisguérin
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
24
|
Zaher S, White D, Ridout J, Branco RG, Meyer R, Pathan N. The effect of nutritional status and inflammatory stimuli on Ghrelin and PYY levels among critically ill children: A prospective and observational study. JPEN J Parenter Enteral Nutr 2022; 46:1298-1306. [DOI: 10.1002/jpen.2339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Sara Zaher
- Clinical Nutrition Department, Faculty of Applied Medical Sciences Taibah University Saudi Arabia
| | - Deborah White
- Department of Paediatrics University of Cambridge, Hills Road Cambridge UK
| | - Jenna Ridout
- Cambridge University Hospitals NHS Foundation Trust, Hills Road Cambridge UK
| | | | - Rosan Meyer
- Department Paediatrics, Imperial Collage London, UK/Department Nutrition and Dietetics, University of Winchester UK
| | - Nazima Pathan
- Department of Paediatrics University of Cambridge, Hills Road Cambridge UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road Cambridge UK
| |
Collapse
|
25
|
Ferrer B, Suresh H, Tinkov AA, Santamaria A, Rocha JB, Skalny AV, Bowman AB, Aschner M. Ghrelin attenuates methylmercury-induced oxidative stress in neuronal cells. Mol Neurobiol 2022; 59:2098-2115. [PMID: 35040042 DOI: 10.1007/s12035-022-02726-5] [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: 11/02/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022]
Abstract
Methylmercury (MeHg) is a global pollutant, which can cause damage to the central nervous system at both high-acute and chronic-low exposures, especially in vulnerable populations, such as children and pregnant women. Nowadays, acute-high poisoning is rare. However, chronic exposure to low MeHg concentrations via fish consumption remains a health concern. Current therapeutic strategies for MeHg poisoning are based on the use of chelators. However, these therapies have limited efficacy. Ghrelin is a gut hormone with an important role in regulating physiologic processes. It has been reported that ghrelin plays a protective role against the toxicity of several xenobiotics. Here, we explored the role of ghrelin as a putative protector against MeHg-induced oxidative stress. Our data show that ghrelin was able to ameliorate MeHg-induced reactive oxygen species (ROS) production in primary neuronal hypothalamic and hippocampal cultures. An analogous effect was observed in mouse hypothalamic neuronal GT 1-7 cells. Using this model, our novel findings show that antioxidant protection of ghrelin against MeHg is mediated by glutathione upregulation and induction of the NRF2/NQO1 pathway.
Collapse
Affiliation(s)
- Beatriz Ferrer
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| | - Harshini Suresh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.,Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, Russia
| | - Abel Santamaria
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular Y Nanotecnología, Instituto Nacional de Neurología Y Neurocirugía, 14269, Mexico City, Mexico
| | - João Batista Rocha
- Departamento de Bioquímica E Biologia Molecular, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Anatoly V Skalny
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", IM Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia.,Department of Bioelementology, KG Razumovsky Moscow State University of Technologies and Management, Moscow, Russia
| | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA. .,Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
26
|
Hu Z, Zhang T, Mei Y, Sun N, Lv K, Wang D. Impact of Ghrelin on Ventricular Arrhythmia and Related Mechanism After Myocardial Infarction. Pharmacology 2021; 107:102-110. [PMID: 34718242 DOI: 10.1159/000519330] [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: 12/12/2020] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Ghrelin is an endogenous peptide with potential protective effects on ischemic heart. METHODS Synthetic ghrelin was administered (100 μg·kg-1 subcutaneous injection, twice daily) for 4 weeks in a rat model of myocardial infarction (MI) with coronary artery occlusion. At the 5th week, electrocardiogram, monophasic action potentials and autonomic nerve function were evaluated. Cardiac tyrosine hydroxylase (TH) was determined by immunofluorescence staining. RESULTS MI significantly increased sympathetic nerve activity (SNA) and ventricular arrhythmias, and prolonged APD dispersion and APD alternans (p < 0.01). Ghrelin treatment significantly increased ventricular fibrillation threshold (VFT), shortened APD dispersion and APD alternans, inhibited SNA and promoted vagus nerve activities (p < 0.01). Ghrelin also markedly reversed abnormal expression of TH in the peri-infarcted area of the heart (p < 0.01). DISCUSSION/CONCLUSION Ghrelin provides a sustained electrophysiological protection by the increase of VFT and improvement of APD dispersion and APD alternans. The mechanism may be related to the regulation of autonomic nerve and sympathetic nerve remodeling. Thus, ghrelin represents a novel drug to prevent ventricular arrhythmia in ischemic heart disease.
Collapse
Affiliation(s)
- Zhengtao Hu
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Ting Zhang
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
- Department of Psychology, Wannan Medical College, Wuhu, China
| | - Yong Mei
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Nan Sun
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
| | - Deguo Wang
- Department of Gerontology, First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
| |
Collapse
|
27
|
Genkwanin suppresses MPP +-induced cytotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in a cellular model of Parkinson's disease. Neurotoxicology 2021; 87:62-69. [PMID: 34481870 DOI: 10.1016/j.neuro.2021.08.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/09/2021] [Accepted: 08/31/2021] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is a complicated multifactorial neurodegenerative disorder. Oxidative stress, neuroinflammatory response, and activation of apoptosis have been proposed to be tightly involved in the pathogenesis of PD. Genkwanin is a typical bioactive non-glycosylated flavonoid with anti-inflammatory and anti-oxidant activities. However, the effect of genkwanin on PD remains unclear. Cell viability, lactate dehydrogenase (LDH) release, caspase-3/7 activity, and apoptosis was evaluated by MTT, LDH release assay, caspase-3/7 activity assay, and TUNEL assay, respectively. The secretion of prostaglandin E2 (PGE2), tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 were measured by respective commercial ELISA kits. The mRNA expression of TNF-α, IL-1β, and IL-6 was detected by qRT-PCR. The protein levels of cycloxygenase-2 (COX-2), toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and NOD-like receptor (NLR) protein: 3 (NLRP3) were determined by western blot analysis. Genkwanin at concentrations less than 40 μM had no impact on cell viability and LDH release. Genkwanin suppressed MPP+-induced neuroinflammation in SH-SY5Y cells. MPP+ treatment inhibited cell viability, increased LDH release, apoptosis, and ROS generation, and reduced superoxide dismutase (SOD) activity in SH-SY5Y cells, which were abolished by genkwanin treatment. Genkwanin suppressed MPP+-induced activation of TLR4/MyD88/NLRP3 inflammasome pathway in SH-SY5Y cells. TLR4 overexpression weakened the anti-inflammatory and anti-neurotoxicity of genkwanin in SH-SY5Y cells. In conclusion, genkwanin attenuated neuroinflammation and neurotoxicity by inhibiting TLR4/MyD88/NLRP3 inflammasome pathway in MPP+-induced cellular model of PD.
Collapse
|
28
|
Hu Y, Ye C, Cheng S, Chen J. Propofol Downregulates lncRNA MALAT1 to Alleviate Cerebral Ischemia-Reperfusion Injury. Inflammation 2021; 44:2580-2591. [PMID: 34427851 DOI: 10.1007/s10753-021-01525-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 10/20/2022]
Abstract
Propofol (PPF) is reported to play a protective role in ischemia/reperfusion (I/R) injury, including cerebral ischemia-reperfusion injury (CIRI). This study aims to investigate the mechanism by which PPF ameliorates CIRI. Kunming mice were used to establish the middle cerebral artery occlusion (MCAO)/reperfusion mouse model in vivo. PPF pre-treatment was performed before CIRI. Lactate dehydrogenase (LDH) and creatine phosphokinase (CPK) levels were detected to evaluate the tissue injury. PC12 cells were exposed to hypoxia/reoxygenation (H/R) to construct the in vitro CIRI model, and PC12 cells were pre-treated with PPF before H/R. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to detect the expression of lncRNA MALAT1 and miR-182-5p. Flow cytometry was used to detect the apoptosis of PC12 cells. Bioinformatics analysis, qRT-PCR, dual-luciferase reporter gene experiments, and RNA immunoprecipitation (RIP) experiments were performed to predict and validate the targeting relationship between MALAT1 and miR-182-5p. Western blot was used to detect Toll-like receptor 4 (TLR4) expression at protein level. PPF pre-treatment remarkably inhibited LDH and CPK levels in the serum of the mice with CIRI, and reduced the apoptosis of PC12 cells exposed to H/R. Besides, PPF pre-treatment markedly suppressed MALAT1 expression in both in vivo and in vitro models and upregulated miR-182-5p expression. MiR-182-5p was validated to be a downstream target gene of MALAT1, and MALAT1 could increase the expression of TLR4 by suppressing miR-182-5p. The effects of PPF on the injury of the mice brain and PC12 cells were partly counteracted by the restoration of MALAT1. PPF protects the brain against I/R-induced injury by regulating MALAT1/miR-182-5p/TLR4 axis.
Collapse
Affiliation(s)
- Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Cong Ye
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Shuang Cheng
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Junyang Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China. .,Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China.
| |
Collapse
|
29
|
Sharma P, Wang X, Ming CLC, Vettori L, Figtree G, Boyle A, Gentile C. Considerations for the Bioengineering of Advanced Cardiac In Vitro Models of Myocardial Infarction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2003765. [PMID: 33464713 DOI: 10.1002/smll.202003765] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Indexed: 06/12/2023]
Abstract
Despite the latest advances in cardiovascular biology and medicine, myocardial infarction (MI) remains one of the major causes of deaths worldwide. While reperfusion of the myocardium is critical to limit the ischemic damage typical of a MI event, it causes detrimental morphological and functional changes known as "reperfusion injury." This complex scenario is poorly represented in currently available models of ischemia/reperfusion injury, leading to a poor translation of findings from the bench to the bedside. However, more recent bioengineered in vitro models of the human heart represent more clinically relevant tools to prevent and treat MI in patients. These include 3D cultures of cardiac cells, the use of patient-derived stem cells, and 3D bioprinting technology. This review aims at highlighting the major features typical of a heart attack while comparing current in vitro, ex vivo, and in vivo models. This information has the potential to further guide in developing novel advanced in vitro cardiac models of ischemia/reperfusion injury. It may pave the way for the generation of advanced pathophysiological cardiac models with the potential to develop personalized therapies.
Collapse
Affiliation(s)
- Poonam Sharma
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Laura Vettori
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Gemma Figtree
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Andrew Boyle
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Carmine Gentile
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| |
Collapse
|
30
|
Coskun ZM, Beydogan AB, Yanar K, Atukeren P, Bolkent S. Oxidative stress and inflammatory response of ghrelin on myocardial and aortic tissues in insulin-resistant rats. J Pharm Pharmacol 2021; 73:692-699. [PMID: 33772291 DOI: 10.1093/jpp/rgab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/29/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES This study was designed to clarify the effects of ghrelin on myocardial and aortic tissues in insulin-resistant rats. METHODS Sprague-Dawley rats were divided into the following groups: control (Group 1), insulin resistance (IR, Group 2), ghrelin (Group 3) and IR+Ghrelin (Group 4) groups. Levels of HOMA-IR, fibronectin, hydroxyproline, collagen-1, collagen-3, matrix metalloproteinase-3, and matrix metalloproteinase-9, and tissue inhibitor of metalloproteinase-1, and oxidative stress parameters as protein carbonyl (PCO), lipid hydroperoxides (LHPs), malondialdehyde, total thiol were determined in myocardial tissue. Expressions of IL-6, NF-κB and TNF-α mRNAs were detected by RT-qPCR. Aorta tissue was stained Masson trichrome. KEY FINDINGS The HOMA-IR level decreased in the IR+Ghrelin group compared with the IR group (P < 0.001). The PCO and LHP concentrations were higher in the IR group compared with control rats (P < 0.05). The PCO level was reduced by ghrelin in the IR+Ghrelin group compared with the IR group (P < 0.001). Ghrelin treatment reduced the mRNA expression levels of IL-6, NF-κB and TNF-α in the IR+Ghrelin group compared with the IR group (P < 0.001). There was no difference among the groups in the histology of aortic tissue. CONCLUSIONS Ghrelin, a regulator of appetite and energy homeostasis, may be effective in regulating oxidative stress and the inflammatory response when impaired by IR. Therefore, ghrelin may reduce the risks of myocardial dysfunction in IR.
Collapse
Affiliation(s)
- Zeynep Mine Coskun
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Demiroglu Bilim University, Istanbul Turkey
| | - Alisa Bahar Beydogan
- Department of Medical Biology, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Karolin Yanar
- Department of Biochemistry, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Pınar Atukeren
- Department of Biochemistry, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sema Bolkent
- Department of Medical Biology, Faculty of Cerrahpasa Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
31
|
Chen H, Du J, Zhang S, Tong H, Zhang M. Ghrelin suppresses migration of macrophages via inhibition of ROCK2 under chronic intermittent hypoxia. J Int Med Res 2021; 48:300060520926065. [PMID: 32485129 PMCID: PMC7273871 DOI: 10.1177/0300060520926065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Objectives Migration of macrophages and atherosclerosis result in various
diseases, including coronary heart disease. This study aimed to
clarify the roles that ghrelin and Rho-associated
coiled-coil-containing protein kinase 2 (ROCK2) play in
migration of macrophages under chronic intermittent hypoxia
(CIH). Methods A rat model of CIH was constructed and changes in ghrelin and ROCK2
protein expression were measured by western blot assay. The
migratory ability of macrophages was determined by the transwell
assay. Hematoxylin and eosin staining was applied to detect the
changes in intima-media thickness. Results We found that CIH enhanced migration of macrophages, and this
effect was attenuated by exogenous ghrelin. Additionally, the
facilitative effect of CIH on migration of macrophages was
strengthened or decreased by upregulation or downregulation of
ROCK2, respectively. This phenomenon indicated that ROCK2 was
involved in CIH-induced migration in macrophages. Furthermore,
western blot and transwell assays showed that ghrelin inhibited
CIH-induced migration via ROCK2 suppression in macrophages. Conclusions In summary, the present study shows that ghrelin inhibits
CIH-induced migration via ROCK2 suppression in macrophages. Our
research may help lead to identifying a new molecular mechanism
for targeted therapy of atherosclerosis and its associated
coronary artery diseases under intermittent hypoxia.
Collapse
Affiliation(s)
- Hong Chen
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, China
| | - Jianfeng Du
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, China
| | - Siying Zhang
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, China
| | - Hao Tong
- China Medical University, Shenyang, China
| | - Man Zhang
- Central Hospital Affiliated to Shenyang Medical College, Shenyang, Liaoning, China
| |
Collapse
|
32
|
Kihara M, Kaiya H, Hirai Y, Katayama H, Terao A, Nishikawa M. Salmon acyl-ghrelin increases food intake and reduces doxorubicin-induced myocardial apoptosis in rats, likely by anti-oxidative activity. Peptides 2021; 137:170471. [PMID: 33340558 DOI: 10.1016/j.peptides.2020.170471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
We had reported that orally administered ghrelin-containing salmon stomach extract prevents doxorubicin (DOX)-induced cardiotoxicity. In this study, we investigated the binding affinity of salmon ghrelin to rat ghrelin receptor and the cardioprotective effects of subcutaneous (sc) injected synthetic salmon ghrelin in rats with DOX-induced acute heart failure in order to clarify the potential efficacy of salmon ghrelin. Intracellular calcium mobilization assay was performed on rat GHS-R1a-expressing CHO cells to reveal ghrelin activity. Rats were divided into five groups; the normal control (I), and toxic control (II) groups were given saline (sc, twice daily), and the salmon acyl-ghrelin (sAG) (III), salmon unacylated-ghrelin (sUAG) (IV), and rat acyl-ghrelin (rAG) (V) groups were given corresponding synthetic ghrelins (sc, twice daily), respectively. After seven days of treatment, DOX (20 mg/kg BW) or saline was administered to the corresponding groups by intraperitoneal injection. The toxic control group was the negative control group for the DOX-induced cardiotoxicity groups. While sAG displayed similar affinity to rAG upon application to GHS-R1a-expressing cells, and also decreased DOX-induced apoptosis and increased food intake, sUAG did not. Both sAG and rAG improved DOX-induced deterioration, showing anti-oxidative activity. The anti-oxidative activity of sAG might contribute to the protective effects on cardiomyocytes. The results also suggest that, similar to rAG, sAG is a potent protectant against DOX-induced cardiotoxicity and a potential functional component in orally administered ghrelin-containing salmon stomach extract, which prevented DOX-induced cardiotoxicity in our previous study.
Collapse
Affiliation(s)
- Minoru Kihara
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan.
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, 564-8565, Japan
| | - Yumi Hirai
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Hidekazu Katayama
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| | - Akira Terao
- Liberal Arts Education Center, Sapporo Campus, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Masazumi Nishikawa
- Department of Food Management, School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatadate, Taihaku-ku, Sendai, 982-0215, Japan
| |
Collapse
|
33
|
Shati AA, El-Kott AF. Acylated ghrelin protects against doxorubicin-induced nephropathy by activating silent information regulator 1. Basic Clin Pharmacol Toxicol 2021; 128:805-821. [PMID: 33547742 DOI: 10.1111/bcpt.13569] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 01/01/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
This study investigated the nephroprotective role of acylated ghrelin (AG) against DOX-induced nephropathy and examined whether the protection involves silent information regulator 1 (SIRT1). Rats were divided into control, control + AG, DOX, DOX + AG, DOX + AG + [D-Lys3]-GHRP-6 (a ghrelin receptor antagonist), and DOX + AG + EX-527 (a sirt1 inhibitor). DOX was given over the first 2 weeks. AG (10 ng/kg) and both inhibitors were given as 3 doses/wk for 5 weeks. AG improved the structure and the function of the kidneys; down-regulated the renal expression of TGF-β1, collagen 1A1 and α-SMA; and inhibited the renal collagen deposition in the kidneys of DOX-treated rats. Concomitantly, it reduced the renal levels of ROS, MDA, TNF-α, and IL-6 and protein levels of cytochrome-c, TGF-β1, Smad3 and α-SMA in these rats. In both the control and DOX-treated rats, AG significantly increased the renal levels of SOD and GSH, decreased the expression of cleaved caspase-3 and Bax, increased the total levels and the nuclear activity of SIRT1 and reduced the deacetylation of p53, NF-κB and FOXO-31. All the effects were abolished by the concurrent administration of EX-527 and [D-Lys3]-GHRP-6. In conclusion, AG prevents DOX-induced nephropathy in SIRT1 and GSHRa1-dependent mechanism.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| |
Collapse
|
34
|
Wang H, Dou S, Zhu J, Shao Z, Wang C, Cheng B. Regulatory effects of ghrelin on endoplasmic reticulum stress, oxidative stress, and autophagy: Therapeutic potential. Neuropeptides 2021; 85:102112. [PMID: 33333485 DOI: 10.1016/j.npep.2020.102112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a regulatory peptide that is the endogenous ligand of the growth hormone secretagogue 1a (GHS-R1a) which belongs to the G protein-coupled receptor family. Ghrelin and GHS-R1a are widely expressed in the central and peripheral tissues and play therapeutic potential roles in the cytoprotection of many internal organs. Endoplasmic reticulum stress (ERS), oxidative stress, and autophagy dysfunction, which are involved in various diseases. In recent years, accumulating evidence has suggested that ghrelin exerts protective effects by regulating ERS, oxidative stress, and autophagy in diverse diseases. This review article summarizes information about the roles of the ghrelin system on ERS, oxidative stress, and autophagy in multiple diseases. It is suggested that ghrelin positively affects the treatment of diseases and may be considered as a therapeutic drug in many illnesses.
Collapse
Affiliation(s)
- Huiqing Wang
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Shanshan Dou
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Junge Zhu
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Ziqi Shao
- Cheeloo College of Medicine, Shandong University, 250014 Jinan, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, 272067 Jining, China
| | - Baohua Cheng
- Neurobiology Institute, Jining Medical University, 272067 Jining, China.
| |
Collapse
|
35
|
Yu P, Li Y, Fu W, Li X, Liu Y, Wang Y, Yu X, Xu H, Sui D. Panax quinquefolius L. Saponins Protect Myocardial Ischemia Reperfusion No-Reflow Through Inhibiting the Activation of NLRP3 Inflammasome via TLR4/MyD88/NF-κB Signaling Pathway. Front Pharmacol 2021; 11:607813. [PMID: 33628178 PMCID: PMC7898550 DOI: 10.3389/fphar.2020.607813] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/11/2020] [Indexed: 12/24/2022] Open
Abstract
At present, many patients who undergo reperfusion immediately after percutaneous coronary intervention will undergo microvascular obstruction and reduction in myocardial blood flow. This phenomenon is called "no-reflow (NR)," and there is still no effective therapy for NR. Studies showed Panax quinquefolius L. saponins (PQS) have effect on MI/R injury, while the effect and mechanism of PQS on MI/R induced NR are not clear. In this study, we established a MI/R model to investigate whether PQS decrease NR phenomenon via suppression of inflammation. We found that PQS significantly alleviated the symptoms of NR by reducing ischemia, infarction, and NR area; improving cardiac function; preventing pathological morphology changes of myocardium; depressing leukocytes' aggregation and adhesion; and suppressing the excessive inflammation. Further study demonstrated that PQS remarkably inhibited TLR4, MyD88, p-NF-κB, and NLRP3 inflammasome-associated protein, and these effects could be reversed by LPS. These results indicated that PQS may protect NR by inhibiting the activation of NLRP3 inflammasome via TLR4/MyD88/NF-κB signaling pathway in part, suggesting that PQS exist potential in preventing NR induced by MI/R.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
36
|
Yuan X, Li S, Zhou L, Tang T, Cheng Y, Ao X, Tan L. Diagnostic Accuracy of Plasma Ghrelin Concentrations in Pediatric Sepsis-Associated Acute Respiratory Distress Syndrome: A Single-Center Cohort Study. Front Pediatr 2021; 9:664052. [PMID: 34095029 PMCID: PMC8175974 DOI: 10.3389/fped.2021.664052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Ghrelin is the endogenous ligand of growth hormone secretagogue receptor 1a, which plays a role in regulating immunity and inflammation. The aim of this study is to assess the diagnostic value of plasma ghrelin in sepsis-associated pediatric acute respiratory distress syndrome (PARDS). Methods: We recruited patients who were admitted to the pediatric ICU (PICU) of the Children's Hospital of Chongqing Medical University between January 2019 and January 2020 and met the diagnostic criteria for sepsis. Data on clinical variables, laboratory indicators, plasma ghrelin concentrations, and inflammatory factors were collected and evaluated, and patients were followed up for 28 days. The area under the receiver-operating characteristic curves (AUROC) were determined using logistic regression to calculate and test cut-off values for ghrelin as a diagnostic indicator of sepsis-associated PARDS. The log-rank test was used to compare survival according to ghrelin levels. Main results: Sixty-six PICU patients (30 with ARDS and 36 without ARDS) who met the diagnostic criteria of sepsis were recruited. The ghrelin level was significantly higher in the ARDS group than in the non-ARDS group. The AUROC of ghrelin was 0.708 (95% confidence interval: 0.584-0.833) and the positivity cutoff value was 445 pg/mL. Sensitivity, specificity, positive predictive value, negative predictive value, positive likelihood ratio, and negative likelihood ratio of plasma ghrelin for the diagnosis of PARDS-associated sepsis were 86.7, 50.0, 59.1, 81.8, 1.734, and 0.266%, respectively. The survival rate of sepsis patients were significantly improved when the ghrelin level was >445 pg/mL. Conclusions: Ghrelin plasma levels were higher in sepsis-associated PARDS, and accompanied by increased levels of inflammatory factors. High ghrelin levels are a positive predictor of ICU survival in sepsis patients. Yet, there is no evidence to prove that elevated ghrelin is a promising diagnostic indicator of sepsis-associated PARDS. Trial registration: Clinicaltrials, ChiCTR1900023254. Registered 1 December 2018 - Retrospectively registered, http://www.clinicaltrials.gov/ChiCTR1900023254.
Collapse
Affiliation(s)
- Xiu Yuan
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Shaojun Li
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders (Chongqing), Chongqing, China
| | - Liang Zhou
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Tian Tang
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuwei Cheng
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Xiaoxiao Ao
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liping Tan
- Department of Emergency, Children's Hospital of Chongqing Medical University, Chongqing, China.,National Clinical Research Center for Child Health and Disorders (Chongqing), Chongqing, China
| |
Collapse
|
37
|
Zhang Y, Zhao Q. AEG-1 deletion promotes cartilage repair and modulates bone remodeling-related cytokines via TLR4/MyD88/NF-κB inhibition in ovariectomized rats with osteoporosis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1298. [PMID: 33209878 PMCID: PMC7661885 DOI: 10.21037/atm-20-5842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background Osteoporosis is a systemic skeletal disorder that can impact a variety of bones throughout the body. Astrocyte-elevated gene-1 (AEG-1) is involved in multiple pro-tumorigenic functions and participates in various inflammatory reactions. However, whether it has an impact on osteoporosis-related cartilage repair and bone remodeling remains unknown. Methods We utilized an ovariectomy mouse model with AEG-1 deletion to investigate the role of AEG-1 in osteoporosis. The mRNA level of AEG-1 was detected by RT-PCR, bone markers, bone volume/total volume (BV/TV), trabecular bone surface/bone volume (BSA/BV) and trabecular bone thickness (Tb. Th) were detected by micro computed tomography (µCT), bone injury was observed by HE and alcian blue staining. The contents of IL-6, IL-17, iNOS and IL-10 in peripheral blood of the three groups were detected by ELISA. The expression of OSX, coi1a1, OC, TLR4, MyD88 and NF-κB were detected by Western Blot. Results µCT revealed increased bone volume in the AEG-1 knockout (KO) ovariectomy (OVX) group compared to the wildtype (WT) OVX group 4 weeks after surgery, indicating restored bone formation after AEG-1 deletion. Flow sorting revealed that AEG-1 deletion inhibited the production of inflammatory factors. Western blot demonstrated activation of the TLR4/MyD88/NF-κB pathway after LPS exposure, which was reduced by AEG-1 deletion. AEG-1 deletion also improved lipopolysaccharide (LPS) induced adverse reactions. Conclusions Taken together, these findings indicate that AEG-1 deletion improves cartilage repair and bone remodeling during osteoporosis, which may partly occur through the inhibition of the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Rheumatology, Henan University Huai He Hospital, Kaifeng City, China
| | - Qing Zhao
- Department of Rheumatology, Henan University Huai He Hospital, Kaifeng City, China
| |
Collapse
|
38
|
Regional Differences in the Ghrelin-Growth Hormone Secretagogue Receptor Signalling System in Human Heart Disease. CJC Open 2020; 3:182-194. [PMID: 33644732 PMCID: PMC7893201 DOI: 10.1016/j.cjco.2020.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Background The hormone ghrelin and its receptor, the growth hormone secretagogue receptor (GHSR) are expressed in myocardium. GHSR binding activates signalling pathways coupled to cardiomyocyte survival and contractility. These properties have made the ghrelin-GHSR axis a candidate for a biomarker of cardiac function. The dynamics of ghrelin-GHSR are altered significantly in late stages of heart failure (HF) and cardiomyopathy, when left ventricular (LV) function is failing. We examined the relationship of GHSR with ghrelin in cardiac tissue from patients with valvular disease with no detectable changes in LV function. Methods Biopsy samples from the left ventricle and left atrium were obtained from 25 patients with valvular disease (of whom 13 also had coronary artery disease) and preserved LV ejection fraction, and compared to control samples obtained via autopsy. Using quantitative confocal fluorescence microscopy, levels of GHSR were determined using [Dpr3(n-octanoyl),Lys19(sulfo-Cy5)]ghrelin(1-19), and immunofluorescence determined ghrelin, the heart failure marker natriuretic peptide type-B (BNP), and contractility marker sarcoplasmic reticulum ATPase pump (SERCA2a). Results A positive correlation between GHSR and ghrelin was apparent in only diseased tissue. Ghrelin and BNP significantly correlated in the left ventricle and strongly colocalized to the same intracellular compartment in diseased and control tissue. GHSR, ghrelin, and BNP all strongly and significantly correlated with SERCA2a in the left ventricle of diseased tissue only. Conclusions Our results suggest that the dynamics of the myocardial ghrelin-GHSR axis is altered in cardiovascular disease in the absence of measurable changes in heart function, and might accompany a regional shift in endocrine programming.
Collapse
|
39
|
Gupta S, Mitra A. Heal the heart through gut (hormone) ghrelin: a potential player to combat heart failure. Heart Fail Rev 2020; 26:417-435. [PMID: 33025414 DOI: 10.1007/s10741-020-10032-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor-mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor-mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman, 713347, India
| | - Arkadeep Mitra
- Department of Zoology, City College , 102/1, Raja Rammohan Sarani, Kolkata, 700009, India.
| |
Collapse
|
40
|
Yu Y, Xu Z, Zhang Y, Wang Q, Ou C, Wang Y, Wang L, Gao P, Du S, Guo F, Ma J. Ghrelin attenuates infectious bursal disease virus-induced early inflammatory response and bursal injury in chicken. Poult Sci 2020; 99:5399-5406. [PMID: 33142456 PMCID: PMC7647848 DOI: 10.1016/j.psj.2020.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 07/03/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023] Open
Abstract
Studies demonstrated that chicken ghrelin mRNA was expressed in immune organs of chicken. However, it was not known for its functions in chicken immune system. This study aimed to investigate the effects of ghrelin on infectious bursal disease virus (IBDV)-induced acute inflammatory and bursal injury. Chickens were divided into 4 groups. One group was used as control (“C”). The other three groups incubated with IBDV on the 19th d, of which 2 were injected intraperitoneally with 0.5 nmol (“LG”) or 1.0 nmol (“HG”) ghrelin/100g body weight from 18th to 22nd d, respectively, and one was injected intraperitoneally with PBS (“I”). Results showed that cytokines including interleukin (IL)-6, IL-1β, and IL-8 mRNA expression in I group were upregulated significantly after chickens infected with IBDV from 1 d post-infection (dpi) to 3 dpi (P < 0.05). However, the expression level of IL-6, IL-1β, and IL-8 mRNA in LG and HG groups was 7.3, ∼43.3% as much as that of the I group at 2 dpi and 3 dpi (P < 0.05). Moreover, ghrelin administration attenuated significantly the bursal injury from 1 dpi to 7 dpi and prevents the reduction of bird weight gain at 5 dpi and 7 dpi, which were induced by IBDV (P < 0.05). The results indicated that ghrelin could play an important role in the immune system of chicken.
Collapse
Affiliation(s)
- Yan Yu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Zhiyong Xu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yanhong Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Qiuxia Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Changbo Ou
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yimin Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Li Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Pei Gao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Shouyang Du
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Feng Guo
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Jinyou Ma
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China.
| |
Collapse
|
41
|
Chen QF, Wang W, Huang Z, Huang DL. Hypoxia-inducible factor-1α attenuates myocardial inflammatory injury in rats induced by coronary microembolization. AN ACAD BRAS CIENC 2020; 92:e20190658. [PMID: 32428089 DOI: 10.1590/0001-3765202020190658] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
To investigated the role of HIF-1α in myocardial inflammatory injury in rats induced by CME and its possible mechanism. Forty SD rats were separated randomly and equally into four groups, i.e. CME+HIF-1α stabilizer dimethyloxalyl glycine (CME+DMOG) group, CME+HIF-1α inhibitor YC-1 (CME+YC-1) group, CME group, and Sham group. HBFP staining, myocardial enzyme assessment, and cardiac ultrasound were used to measure microinfarct, serum c-troponin I (cTnI) level, and Cardiac function. ELISA and western blot were applied for detecting NLRP3 inflammasome pathway and TLR4/MyD88/NF-κB signaling level.Pro-inflammatory factors of IL-18, IL-1β and TNF-α increased their expression levels after CME, which indicated inflammatory responses in the myocardium. Additionally, in the inflammatory process, NLRP3 inflammasome and TLR4/MyD88/NF-κB signaling were involved. DMOG reverses these effects of CME, whereas YC-1 aggravates these effects. HIF-1α may attenuate myocardial inflammatory injury induced by CME and improve cardiac function, which can perhaps be explained by the fact that TLR4/MyD88/NF-κB signaling pathway activation is inhibited.
Collapse
Affiliation(s)
- Quan-Fang Chen
- the First Affiliated Hospital, Guangxi Medical University, Guangxi, China
| | - Wei Wang
- the First Affiliated Hospital, Guangxi Medical University, Guangxi, China
| | - Zhou Huang
- the First Affiliated Hospital, Guangxi Medical University, Guangxi, China
| | - Dong-Ling Huang
- the First Affiliated Hospital, Guangxi Medical University, Guangxi, China
| |
Collapse
|
42
|
Liu Y, Xu XY, Shen Y, Ye CF, Hu N, Yao Q, Lv XZ, Long SL, Ren C, Lang YY, Liu YL. Ghrelin protects against obesity-induced myocardial injury by regulating the lncRNA H19/miR-29a/IGF-1 signalling axis. Exp Mol Pathol 2020; 114:104405. [PMID: 32084395 DOI: 10.1016/j.yexmp.2020.104405] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 01/10/2020] [Accepted: 02/15/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Obesity is associated with the impairment of cardiac fitness and consequent ventricular dysfunction and heart failure. Ghrelin has been largely documented to be cardioprotective against ischaemia/reperfusion injury. However, the role of ghrelin in obesity-induced myocardial injury is largely unknown. This study sought to determine the cardiac effect of ghrelin against obesity-induced injury and the underlying mechanisms. METHODS The effect of ghrelin was evaluated in a mouse model of obesity and a palmitic acid (PA)-treated cardiomyocyte cell line with or without ghrelin transfection. Gene and protein expression levels were determined by real-time PCR and western blot, respectively. Cell apoptosis was measured by flow cytometry analysis. RESULTS In the present study, we found that both a high-fat diet (HFD) and PA treatment caused myocardial injury by increasing apoptosis and the expression of inflammatory cytokines. Overexpression of ghrelin reversed the effects induced by HFD or PA treatment. Knockdown of lncRNA H19 or overexpression of miR-29a abrogated the cardioprotective effects of ghrelin against apoptosis and inflammation. We also found that IGF-1 was a target gene of miR-29a and that H19 regulated IGF-1 expression via miR-29a. Overexpression of IGF-1 partially reversed the apoptosis and inflammation promoting effects of miR-29a. CONCLUSIONS Our findings suggested that ghrelin protected against obesity-induced myocardial injury by regulating the H19/miR-29a/IGF-1 signalling axis, providing further evidence for the clinical application of ghrelin.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Xin-Yue Xu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yang Shen
- Molecular medicine laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Chun-Feng Ye
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Na Hu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Qing Yao
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Xiu-Zi Lv
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Sheng-Lan Long
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Chao Ren
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yuan-Yuan Lang
- Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| | - Yan-Ling Liu
- Department of Pediatrics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
43
|
Femminò S, Pagliaro P, Penna C. Obesity and Cardioprotection. Curr Med Chem 2020; 27:230-239. [DOI: 10.2174/0929867326666190325094453] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/05/2018] [Accepted: 12/24/2018] [Indexed: 01/21/2023]
Abstract
The incidence of obesity and diabetes is increasing rapidly worldwide. Obesity and
metabolic syndrome are strictly linked and represent the basis of different cardiovascular risk
factors, including hypertension and inflammatory processes predisposing to ischemic heart
disease, which represent the most common causes of heart failure. Recent advances in the understanding
of ischemia/reperfusion mechanisms of injury and mechanisms of cardioprotection
are briefly considered. Resistance to cardioprotection may be correlated with the severity
of obesity. The observation that heart failure obese patients have a better clinical condition
than lean heart failure patients is known as “obesity paradox”. It seems that obese patients
with heart failure are younger, making age the most important confounder in some studies.
Critical issues are represented by the "obesity paradox” and heart failure exacerbation by inflammation.
For heart failure exacerbation by inflammation, an important role is played by
NLRP3 inflammasome, which is emerging as a possible target for heart failure condition.
These critical issues in the field of obesity and cardiovascular diseases need more studies to
ascertain which metabolic alterations are crucial for alleged beneficial and deleterious effects
of obesity.
Collapse
Affiliation(s)
- Saveria Femminò
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
44
|
Cheng Y, Chen B, Xie W, Chen Z, Yang G, Cai Y, Shang H, Zhao W. Ghrelin attenuates secondary brain injury following intracerebral hemorrhage by inhibiting NLRP3 inflammasome activation and promoting Nrf2/ARE signaling pathway in mice. Int Immunopharmacol 2020; 79:106180. [PMID: 31926478 DOI: 10.1016/j.intimp.2019.106180] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/16/2019] [Accepted: 12/31/2019] [Indexed: 02/08/2023]
Abstract
Ghrelin, a brain-gut peptide, has been proven to exert neuroprotection in different kinds of neurological diseases; however, its role and the potential molecular mechanisms in secondary brain injury (SBI) after intracerebral hemorrhage (ICH) are still unknown. In this study, we investigate whether treatment with ghrelin may attenuate SBI in a murine ICH model, and if so, whether the neuroprotective effects are due to the inhibition of nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome activation and promotion of nuclear factor-E2-related factor 2 (Nrf2)/antioxidative response element (ARE) signaling pathway. Stereotactically intrastriatal infusion of autologous blood was performed to mimic ICH. Ghrelin was given intraperitoneally immediately following ICH and again 1 h later. Results showed that ghrelin attenuated neurobehavioral deficits, brain edema, hematoma volume, and perihematomal cell death post-ICH. Ghrelin inhibited the NLRP3 inflammasome activation and subsequently suppressed the neuroinflammatory response as evidenced by reduced microglia activation, neutrophil infiltration, and pro-inflammatory mediators release after ICH. Additionally, ghrelin alleviated ICH-induced oxidative stress according to the chemiluminescence of luminol and lucigenin, malondialdehyde (MDA) content, and total superoxide dismutase (SOD) activity assays. These changes were accompanied by upregulation of Nrf2 expression, Nrf2 nuclear accumulation, and enhanced Nrf2 DNA binding activity, as well as by increased expressions of Nrf2 downstream target antioxidative genes, including NAD(P)H quinine oxidoreductase-1 (NQO1), glutathione cysteine ligase regulatory subunit (GCLC), and glutathione cysteine ligase modulatory subunit (GCLM). Together, our data suggested that ghrelin protected against ICH-induced SBI by inhibiting NLRP3 inflammasome activation and promoting Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Yijun Cheng
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Bin Chen
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Wanqun Xie
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Zhenghong Chen
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Guoyuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, PR China; Department of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yu Cai
- Department of Neurosurgery, North Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hanbing Shang
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Weiguo Zhao
- Department of Neurosurgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
45
|
Shao XF, Li B, Shen J, Wang QF, Chen SS, Jiang XC, Qiang D. Ghrelin alleviates traumatic brain injury-induced acute lung injury through pyroptosis/NF-κB pathway. Int Immunopharmacol 2020; 79:106175. [PMID: 31918060 DOI: 10.1016/j.intimp.2019.106175] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/22/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022]
Abstract
Acute lung injury (ALI) is one of the severe complications in patients with traumatic brain injury (TBI), contributing to the high mortality. Ghrelin has protective effects against various inflammatory diseases, but the effects of Ghrelin on TBI-induced ALI and its mechanisms remain unknown. In this study, Ghrelin administration was performed on the mice with TBI, then histological change in cortex and lung tissues, lung vascular permeability and macrophage number in bronchoalveolar lavage fluid (BALF) were examined, respectively. Simultaneously, the alterations of proinflammatory factors and pyroptosis-related proteins in lung tissues were detected. As a result, TBI-induced ALI was ameliorated after Ghrelin treatment, which was demonstrated by improved histology, reduced lung vascular permeability, and peripheral macrophage number. Furthermore, Ghrelin decreased the mRNA levels of proinflammatory factors (IL-1β, IL-6, TNF-α and IL-18), the protein levels of pyroptosis-related proteins (NLRP3, Caspase1-P20, HMGB1 and Gasdermin D), and the phosphorylation levels of NF-κB in lung tissues. These results showed that Ghrelin attenuating TBI-induced ALI might be via ameliorating inflammasome-induced pyroptosis by blocking NF-κB signal, which are important for the prevention and treatment of TBI-induced ALI.
Collapse
Affiliation(s)
- Xue-Fei Shao
- Department of Neurosurgery, Yi Ji Shan Hospital of Wannan Medical College, Wuhu, China.
| | - Bo Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jun Shen
- Department of Neurosurgery, Yi Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Qi-Fu Wang
- Department of Neurosurgery, Yi Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - San-Song Chen
- Department of Neurosurgery, Yi Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Xiao-Chun Jiang
- Department of Neurosurgery, Yi Ji Shan Hospital of Wannan Medical College, Wuhu, China
| | - Di Qiang
- Department of Dermatology and STD, Yi-Ji Shan Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
46
|
Biemmi V, Milano G, Ciullo A, Cervio E, Burrello J, Dei Cas M, Paroni R, Tallone T, Moccetti T, Pedrazzini G, Longnus S, Vassalli G, Barile L. Inflammatory extracellular vesicles prompt heart dysfunction via TRL4-dependent NF-κB activation. Theranostics 2020; 10:2773-2790. [PMID: 32194834 PMCID: PMC7052909 DOI: 10.7150/thno.39072] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Background: After myocardial infarction, necrotic cardiomyocytes release damage-associated proteins that stimulate innate immune pathways and macrophage tissue infiltration, which drives inflammation and myocardial remodeling. Circulating inflammatory extracellular vesicles play a crucial role in the acute and chronic phases of ischemia, in terms of inflammatory progression. In this study, we hypothesize that the paracrine effect mediated by these vesicles induces direct cytotoxicity in cardiomyocytes. Thus, we examined whether reducing the generation of inflammatory vesicles within the first few hours after the ischemic event ameliorates cardiac outcome at short and long time points. Methods: Myocardial infarction was induced in rats that were previously injected intraperitoneally with a chemical inhibitor of extracellular-vesicle biogenesis. Heart global function was assessed by echocardiography performed at 7, 14 and 28 days after MI. Cardiac outcome was also evaluated by hemodynamic analysis at sacrifice. Cytotoxic effects of circulating EV were evaluated ex-vivo in a Langendorff, system by measuring the level of cardiac troponin I (cTnI) in the perfusate. Mechanisms undergoing cytotoxic effects of EV derived from pro-inflammatory macrophages (M1) were studied in-vitro in primary rat neonatal cardiomyocytes. Results: Inflammatory response following myocardial infarction dramatically increased the number of circulating extracellular vesicles carrying alarmins such as IL-1α, IL-1β and Rantes. Reducing the boost in inflammatory vesicles during the acute phase of ischemia resulted in preserved left ventricular ejection fraction in vivo. Hemodynamic analysis confirmed functional recovery by displaying higher velocity of left ventricular relaxation and improved contractility. When added to the perfusate of isolated hearts, post-infarction circulating vesicles induced significantly more cell death in adult cardiomyocytes, as assessed by cTnI release, comparing to circulating vesicles isolated from healthy (non-infarcted) rats. In vitro inflammatory extracellular vesicles induce cell death by driving nuclear translocation of NF-κB into nuclei of cardiomyocytes. Conclusion: Our data suggest that targeting circulating extracellular vesicles during the acute phase of myocardial infarction may offer an effective therapeutic approach to preserve function of ischemic heart.
Collapse
Affiliation(s)
- Vanessa Biemmi
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Giuseppina Milano
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Dept. Cœur-Vaisseaux, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Alessandra Ciullo
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Elisabetta Cervio
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Jacopo Burrello
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Michele Dei Cas
- Department of Health Sciences of the University of Milan, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences of the University of Milan, Milan, Italy
| | - Tiziano Tallone
- Cell and Biomedical Technologies Unit Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Tiziano Moccetti
- Department of Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland
| | - Giovanni Pedrazzini
- Department of Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Sarah Longnus
- Department of Cardiovascular Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Giuseppe Vassalli
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation, Lugano, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
47
|
Chen Y, Wang H, Zhang Y, Wang Z, Liu S, Cui L. Pretreatment of ghrelin protects H9c2 cells against hypoxia/reoxygenation-induced cell death via PI3K/AKT and AMPK pathways. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2179-2187. [PMID: 31159591 DOI: 10.1080/21691401.2019.1620253] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ghrelin has been widely recognized as a key peptide in the cardiovascular system. This study detected the potential of ghrelin in MI management and tried to decode one of the possible underlying mechanisms. H9c2 cells were pretreated with ghrelin and were subjected to hypoxia/reoxygenation (H/R). CCK-8, flow cytometry, Western blot and LDH analysis were conducted to assess the changes in cell survival. LY294002 and Compound C were used to treat H9c2 cells for blocking PI3K/AKT and AMPK pathways, respectively. Ghrelin expression in H9c2 cells was suppressed by siRNA-mediated silencing to see the effects of endogenous ghrelin. We found that, following H/R, H9c2 cells viability was decreased, CyclinD1 and CDK4 were down-regulated, apoptosis was induced, the release of LDH was enhanced, and the expression levels of Cox-2 and iNOS were up-regulated. Ghrelin protected H9c2 cells against H/R induced these alterations. Besides, ghrelin activated PI3K/AKT and AMPK pathways even in H/R-stimulated cells. The protective effects of ghrelin against H/R-induced cell damage were all attenuated by the addition of LY294002 or Compound C. Moreover, endogenous inhibition of ghrelin significantly induced cell death of H9c2 cells. In conclusion, this study demonstrated that ghrelin pretreatment protected H9c2 cells against H/R-induced cell damage, possibly via PI3K/AKT and AMPK pathways.
Collapse
Affiliation(s)
- Yanbo Chen
- a Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , China.,b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Honggang Wang
- c Department of Clinical Laboratory, Weifang People's Hospital , Weifang , Shandong , China
| | - Yong Zhang
- b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Zhengping Wang
- d Department of Ultrasonography, Weifang People's Hospital , Weifang , Shandong , China
| | - Shanshan Liu
- b Department of Cardiology, Weifang People's Hospital , Weifang , Shandong , China
| | - Lianqun Cui
- a Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University , Jinan , Shandong , China
| |
Collapse
|
48
|
Liu F, Li Z, He X, Yu H, Feng J. Ghrelin Attenuates Neuroinflammation and Demyelination in Experimental Autoimmune Encephalomyelitis Involving NLRP3 Inflammasome Signaling Pathway and Pyroptosis. Front Pharmacol 2019; 10:1320. [PMID: 31780940 PMCID: PMC6851267 DOI: 10.3389/fphar.2019.01320] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune and degenerative disease of the central nervous system, and conventional treatments have limited efficacy or side effects. Ghrelin, a 28-amino acid octanoylated peptide, has been reported to have neuroprotective effects, including anti-oxidation, anti-inflammation, and anti-apoptosis. Pyroptosis, also called inflammatory cell death, is triggered by overly active inflammasomes and accompanied by the production of numerous cytokines. As immune dysfunction is primarily involved in the pathogenesis of MS, this study aimed to explore the therapeutic effects and precise functional mechanisms of ghrelin against the nod-like receptor protein 3 (NLRP3) inflammasome and pyroptosis in experimental autoimmune encephalomyelitis (EAE). Sprague Dawley rats were immunized with guinea pig spinal cord homogenates and pertussis toxin to develop an EAE model. All rats were randomly divided into four groups: normal control group, EAE group, EAE + ghrelin group, and ghrelin control group. EAE rats showed abnormal behavioral scores and body weight changes. Histologic analysis displayed severe inflammatory infiltration and demyelination in the brain and spinal cord of EAE rats. Ghrelin treatments potently restored these abnormal changes. In addition, the ghrelin-treated EAE group showed significantly downregulated expression of inflammatory cytokines. The expression of proteins involved in the NLRP3 signaling pathway and pyroptosis was decreased as well. We also found that the anti-inflammatory effect of ghrelin was associated with inhibition of nuclear factor (NF)-κB activation. Compared with rats in the healthy control group, rats in the ghrelin control group did not show statistically significant changes in histologic examinations, pro-inflammatory cytokines production, or molecules involved in the NLRP3 signaling pathway, which indicated that ghrelin induced no side effects in the animals of our study. Our findings provide more insight into the use of ghrelin as a novel candidate for MS.
Collapse
Affiliation(s)
- Fei Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
49
|
Soares ROS, Losada DM, Jordani MC, Évora P, Castro-E-Silva O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int J Mol Sci 2019; 20:ijms20205034. [PMID: 31614478 PMCID: PMC6834141 DOI: 10.3390/ijms20205034] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) permeates a variety of diseases and is a ubiquitous concern in every transplantation proceeding, from whole organs to modest grafts. Given its significance, efforts to evade the damaging effects of both ischemia and reperfusion are abundant in the literature and they consist of several strategies, such as applying pre-ischemic conditioning protocols, improving protection from preservation solutions, thus providing extended cold ischemia time and so on. In this review, we describe many of the latest pharmacological approaches that have been proven effective against IRI, while also revisiting well-established concepts and presenting recent pathophysiological findings in this ever-expanding field. A plethora of promising protocols has emerged in the last few years. They have been showing exciting results regarding protection against IRI by employing drugs that engage several strategies, such as modulating cell-surviving pathways, evading oxidative damage, physically protecting cell membrane integrity, and enhancing cell energetics.
Collapse
Affiliation(s)
| | - Daniele M Losada
- Department of Anatomic Pathology, Faculty of Medical Sciences, University of Campinas, 13083-970 Campinas, Brazil.
| | - Maria C Jordani
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Paulo Évora
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| | - Orlando Castro-E-Silva
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| |
Collapse
|
50
|
Darwesh AM, Sosnowski DK, Lee TYT, Keshavarz-Bahaghighat H, Seubert JM. Insights into the cardioprotective properties of n-3 PUFAs against ischemic heart disease via modulation of the innate immune system. Chem Biol Interact 2019; 308:20-44. [DOI: 10.1016/j.cbi.2019.04.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
|