1
|
Wu Y, Wang X, Zhang J, Du S, Wang Z, Li J, Zhang W, Xiang J, Li R, Liu J, Bi X. Capsaicin combined with stem cells improved mitochondrial dysfunction in PIG3V cells, an immortalized human vitiligo melanocyte cell line, by inhibiting the HSP70/TLR4/mTOR/FAK signaling axis. Mol Biol Rep 2024; 51:650. [PMID: 38734811 DOI: 10.1007/s11033-024-09592-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Vitiligo is a common autoimmune skin disease. Capsaicin has been found to exert a positive effect on vitiligo treatment, and mesenchymal stem cells (MSCs) are also confirmed to be an ideal cell type. This study aimed to explore the influence of capsaicin combined with stem cells on the treatment of vitiligo and to confirm the molecular mechanism of capsaicin combined with stem cells in treating vitiligo. METHODS AND RESULTS PIG3V cell proliferation and apoptosis were detected using CCK-8 and TUNEL assays, MitoSOX Red fluorescence staining was used to measure the mitochondrial ROS level, and JC-1 staining was used to detect the mitochondrial membrane potential. The expression of related genes and proteins was detected using RT‒qPCR and Western blotting. Coimmunoprecipitation was used to analyze the protein interactions between HSP70 and TLR4 or between TLR4 and mTOR. The results showed higher expression of HSP70 in PIG3V cells than in PIG1 cells. The overexpression of HSP70 reduced the proliferation of PIG3V cells, promoted apoptosis, and aggravated mitochondrial dysfunction and autophagy abnormalities. The expression of HSP70 could be inhibited by capsaicin combined with MSCs, which increased the levels of Tyr, Tyrp1 and DCT, promoted the proliferation of PIG3V cells, inhibited apoptosis, activated autophagy, and improved mitochondrial dysfunction. In addition, capsaicin combined with MSCs regulated the expression of TLR4 through HSP70 and subsequently affected the mTOR/FAK signaling pathway CONCLUSIONS: Capsaicin combined with MSCs inhibits TLR4 through HSP70, and the mTOR/FAK signaling pathway is inhibited to alleviate mitochondrial dysfunction and autophagy abnormalities in PIG3V cells.
Collapse
Affiliation(s)
- Yifei Wu
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Xiaochuan Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Jiayu Zhang
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Sha Du
- Department of Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
| | - Zhiqiong Wang
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Jinrong Li
- Department of Dermatology, Traditional Chinese Medicine Hospital of Jinggu County, Pu'er, Yunnan, 666400, China
| | - Wenhe Zhang
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Jie Xiang
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Renfu Li
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Jing Liu
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China
| | - Xin Bi
- Department of Dermatology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming, Yunnan, 650032, China.
| |
Collapse
|
2
|
Shinde A, Shannahan J. Inhalation exposure-induced toxicity and disease mediated via mTOR dysregulation. Exp Biol Med (Maywood) 2024; 249:10135. [PMID: 38711460 PMCID: PMC11070522 DOI: 10.3389/ebm.2024.10135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Environmental air pollution is a global health concern, associated with multiple respiratory and systemic diseases. Epidemiological supports continued urbanization and industrialization increasing the prevalence of inhalation exposures. Exposure to these inhaled pollutants induces toxicity via activation of numerous cellular mechanisms including oxidative stress, autophagy, disrupted cellular metabolism, inflammation, tumorigenesis, and others contributing to disease development. The mechanistic target of rapamycin (mTOR) is a key regulator involved in various cellular processes related to the modulation of metabolism and maintenance of homeostasis. Dysregulation of mTOR occurs following inhalation exposures and has also been implicated in many diseases such as cancer, obesity, cardiovascular disease, diabetes, asthma, and neurodegeneration. Moreover, mTOR plays a fundamental role in protein transcription and translation involved in many inflammatory and autoimmune diseases. It is necessary to understand inhalation exposure-induced dysregulation of mTOR since it is key regulator which may contribute to numerous disease processes. This mini review evaluates the available literature regarding several types of inhalation exposure and their impacts on mTOR signaling. Particularly we focus on the mTOR signaling pathway related outcomes of autophagy, lipid metabolism, and inflammation. Furthermore, we will examine the implications of dysregulated mTOR pathway in exposure-induced diseases. Throughout this mini review, current gaps will be identified related to exposure-induced mTOR dysregulation which may enable the targeting of mTOR signaling for the development of therapeutics.
Collapse
Affiliation(s)
| | - Jonathan Shannahan
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
3
|
Liu X, Qian N, Zhu L, Fan L, Fu G, Ma M, Bao J, Cao C, Liang X. Geniposide ameliorates acute kidney injury via enhancing the phagocytic ability of macrophages towards neutrophil extracellular traps. Eur J Pharmacol 2023; 957:176018. [PMID: 37634840 DOI: 10.1016/j.ejphar.2023.176018] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Acute kidney injury (AKI) is a clinically serious disorder associated with high mortality rates and an increased risk of progression to end-stage renal disease. As an essential supportive treatment for patients with respiratory failure, mechanical ventilation not only save many critically ill patients, but also affect glomerular filtration function by changing renal hemodynamics, neurohumoral and positive end-expiratory pressure, eventually leading to AKI. AMP-activated protein kinase (AMPK), a crucial energy homeostasis regulator, could enhance macrophage phagocytic ability and inhibit inflammation, but whether it can engulf neutrophil extracellular traps (NETs) and alleviate mechanical ventilation-associated AKI is still unclear. In this study, we found that geniposide significantly ameliorated histopathological damage, reduced serum Cre and BUN levels. Besides, geniposide can also induce AMPK activation and enhance macrophage phagocytic ability toward NETs. Moreover, geniposide can markedly reduce the levels of high mobility group box 1 (HMGB1), and these effects were dependent on AMPK-PI3K/Akt signaling. Altogether, these results indicated that geniposide promoted macrophage efferocytosis by inducing AMPK-PI3K/Akt signaling activation, clearing NETs and ameliorating AKI.
Collapse
Affiliation(s)
- Xiaodong Liu
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China; The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Na Qian
- The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Li Zhu
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Li Fan
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China; Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Guanghao Fu
- The Second People's Hospital of Lianyungang, Affiliated to Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
| | - Mengqing Ma
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Jiaxin Bao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China
| | - Changchun Cao
- Department of Nephrology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, Jiangsu, China.
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
4
|
He Y, Wang Y, Yang K, Jiao J, Zhan H, Yang Y, Lv D, Li W, Ding W. Maslinic Acid: A New Compound for the Treatment of Multiple Organ Diseases. Molecules 2022; 27:8732. [PMID: 36557864 PMCID: PMC9786823 DOI: 10.3390/molecules27248732] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Maslinic acid (MA) is a pentacyclic triterpene acid, which exists in many plants, including olive, and is highly safe for human beings. In recent years, it has been reported that MA has anti-inflammatory, antioxidant, anti-tumor, hypoglycemic, neuroprotective and other biological activities. More and more experimental data has shown that MA has a good therapeutic effect on multiple organ diseases, indicating that it has great clinical application potential. In this paper, the extraction, purification, identification and analysis, biological activity, pharmacokinetics in vivo and molecular mechanism of MA in treating various organ diseases are reviewed. It is hoped to provide a new idea for MA to treat various organ diseases.
Collapse
Affiliation(s)
- Yan He
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Yi Wang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Kun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Jia Jiao
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Hong Zhan
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Youjun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - De Lv
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Weihong Li
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| | - Weijun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu 611137, China
| |
Collapse
|
5
|
Zhang W, Ye L, Fang H. Astragaloside IV Improve Neurological Function of Cerebral Ischemia. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study intends to assess astragaloside IV’s effect on neurological function in mice cerebral ischemia model. The mouse model of cerebral ischemia was established by photochemistry and then assigned into sham operation group (photochemical building do not accept cold light
irradiation) and control group (10 ug/ml by intraperitoneal injection of saline solution), drug group (10 ug/ml by intraperitoneal injection of Astragaloside IV) followed by analysis of neurological severity, cerebral infarction area, loss of neurons, glial cell activation and the activities
of LC3, Beclin1, Caspase-3, P62 and mTOR by Western Blot. The neurons in cerebral infarction were missing and marginal area and penumbra appeared. The tissue in cerebral infarction became white, and the modeling was successful. The drug group showed significantly reduced scores and decreased
infarct area of brain tissue compared with control group on day 14, 21 and 28 (P < 0.05). TUNEL staining showed increased number of TUNEL cells at the ischemic edge in the drug group (0.35±0.07)% (P < 0.05), while the IBAL staining of (27.12±3.01)% and GFAP
staining of (0.08±0.02)% in the drug group showed significant inhibition of astrocytes (P < 0.05). The activity of LC3, Beclin1, Caspase-3 and P62 in drug group was inhibited, while the activity of mTOR was promoted. In conclusion, Astragaloside IV improves the balance ability
and the neural function of cerebral ischemia repair in mice model.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Enesthesiology, Enshi Tujia and Miao Autonomous Prefecture Central Hospital, Enshi, Hubei, 445000, China
| | - Lun Ye
- Department of Emergency, Jiangjin Central Hospital of Chongqing, Chongqing, 402260, China
| | - Hairong Fang
- Department of Neurology (II) Ward, The First People’s Hospital of Jiangxia District, Wuhan, Hubei, 430000, China
| |
Collapse
|
6
|
Liang J, Liu J, Tang Y, Peng Q, Zhang L, Ma X, Xu N, Wei J, Han H. Sophoridine inhibits endotoxin‐induced acute lung injury by enhancing autophagy of macrophage and reducing inflammation. J Leukoc Biol 2022; 112:115-125. [PMID: 35603481 DOI: 10.1002/jlb.3ma0322-428r] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 03/06/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jinping Liang
- Infectious disease department,HuaShan Hospital FuDan University Shanghai China
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
- Key Laboratory of Clinical Pathogenic Biology General Hospital of Ningxia Medical University Yinchuan China
| | - Juntong Liu
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Yezhen Tang
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Qian Peng
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Ling Zhang
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Xiaoxia Ma
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Nan Xu
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| | - Jun Wei
- Key Laboratory of Clinical Pathogenic Biology General Hospital of Ningxia Medical University Yinchuan China
| | - Huaiqin Han
- Department of Pathogen Biology and Immunology,School of Basic Medical Sciences Ningxia Medical University Yinchuan750004 China
| |
Collapse
|
7
|
Li L, Lin L, Lei S, Shi S, Chen C, Xia Z. Maslinic Acid Inhibits Myocardial Ischemia-Reperfusion Injury-Induced Apoptosis and Necroptosis via Promoting Autophagic Flux. DNA Cell Biol 2022; 41:487-497. [PMID: 35475713 DOI: 10.1089/dna.2021.0918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Apoptosis, necroptosis, and autophagy are the major programmed cell death in myocardial ischemia-reperfusion injury (MIRI). Maslinic acid (MA) has been found to regulate pathophysiological processes that mediate programmed cell death in MIRI, such as inflammation and oxidative stress. However, its effects on MIRI remain unclear. This study intends to explore the role of MA in MIRI. In vitro, MA had no obvious cytotoxic effects on H9C2 cells, and significantly improved the impaired cell viability caused by hypoxia reoxygenation (HR). In vivo, MA significantly alleviated ischemia reperfusion (IR)-induced left ventricular myocardial tissue injury, downregulated creatine kinase-myocardial band (CK-MB), and lactate dehydrogenase (LDH) levels in serum as well as reducing infarct size. Moreover, MA inhibited HR-induced mitochondrial apoptosis and necroptosis in vitro and in vivo. Of interest, MA interacts with lysosome-associated membrane protein 2 (LAMP2). MA protected LAMP2 from IR and promoting autophagic flux to inhibit apoptosis and necroptosis, whereas these effects were reversed by co-treatment with lysosomal inhibitor BarfA1. In conclusion, MA can inhibit MIRI-induced apoptosis and necroptosis by promoting autophagic flux. These results support that MA is a potential agent to ameliorate MIRI.
Collapse
Affiliation(s)
- Lin Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Lin
- Department of Anesthesiology, YiChang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Si Shi
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chun Chen
- Department of Anesthesiology, YiChang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Li Q, Xu M, Li Z, Li T, Wang Y, Chen Q, Wang Y, Feng J, Yin X, Lu C. Maslinic Acid Attenuates Ischemia/Reperfusion Injury-Induced Myocardial Inflammation and Apoptosis by Regulating HMGB1-TLR4 Axis. Front Cardiovasc Med 2021; 8:768947. [PMID: 34859077 PMCID: PMC8631436 DOI: 10.3389/fcvm.2021.768947] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022] Open
Abstract
Aims: The inflammatory response and apoptosis are the major pathological features of myocardial ischemia/reperfusion injury (MI/RI). Maslinic acid (MA), a natural pentacyclic triterpene with various bioactivities, plays critical roles in the multiple cellular biological processes, but its protective effects on the pathophysiological processes of MI/RI have not been extensively investigated. Our study aimed to determine whether MA treatment alleviate ischemia/reperfusion (I/R)-induced myocardial inflammation and apoptosis both in vitro and in vivo, and further reveal the underlying mechanisms. Methods and results: An MI/RI rat model was successfully established by ligating the left anterior descending coronary artery and H9c2 cells were exposed to hypoxia/reoxygenation (H/R) to mimic I/R injury. In addition, prior to H/R stimulation or myocardial I/R operation, the H9c2 cells or rats were treated with varying concentrations of MA or vehicle for 24 h and two consecutive days, respectively. In this study, our results showed that MA could obviously increase the cell viability and decrease the cardiac enzymes release after H/R in vitro. MA could significantly improve the H/R-induced cardiomyocyte injury and I/R-induced myocardial injury in a dose-dependent manner. Moreover, MA suppressed the expression of inflammatory cytokines (tumor necrosis factor alpha [TNF-α, interleukin-1β [IL-1β and interleukin-6 [IL-6]) and the expressions of apoptosis-related proteins (cleaved caspase-3 and Bax) as well as increased the levels of anti-apoptotic protein Bcl-2 expression both in vitro and in vivo. Mechanistically, MA significantly inhibited nuclear translocation of nuclear factor-κB (NF-κB) p65 after H/R via regulating high mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) axis. Conclusion: Taken together, MA treatment may alleviate MI/RI by suppressing both the inflammation and apoptosis in a dose-dependent manner, and the cardioprotective effect of MA may be partly attributable to the inactivation of HMGB1/TLR4/NF-κB pathway, which offers a new therapeutic strategy for MI/RI.
Collapse
Affiliation(s)
- Qi Li
- School of Medicine, Nankai University, Tianjin, China.,Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Mengping Xu
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Zhuqing Li
- School of Medicine, Nankai University, Tianjin, China.,Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| | - Tingting Li
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Yilin Wang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Qiao Chen
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Yanxin Wang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Jiaxin Feng
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Xuemei Yin
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, China.,Department of Cardiology, The First Center Clinic College of Tianjin Medical University, Tianjin, China
| | - Chengzhi Lu
- School of Medicine, Nankai University, Tianjin, China.,Department of Cardiology, Tianjin First Center Hospital, Tianjin, China
| |
Collapse
|
9
|
Lidocaine Alleviates Sepsis-Induced Acute Lung Injury in Mice by Suppressing Tissue Factor and Matrix Metalloproteinase-2/9. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3827501. [PMID: 34804364 PMCID: PMC8604580 DOI: 10.1155/2021/3827501] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 02/04/2023]
Abstract
Acute lung injury (ALI) is one of the fatal symptoms of sepsis. However, there were no effective clinical treatments. TF accumulation-induced fibrin deposit formations and coagulation abnormalities in pulmonary vessels contribute to the lethality of ALI. Suppressor of cytokine signaling 3 (SOCS3) acts as an endogenous negative regulator of the TLR4/TF pathway. We hypothesized that inducing SOCS3 expression using lidocaine to suppress the TLR4/TF pathway may alleviate ALI. Hematoxylin and eosin (H&E), B-mode ultrasound, and flow cytometry were used to measure the pathological damage of mice. Gelatin zymography was used to measure matrix metalloproteinase-2/9 (MMP-2/9) activities. Western blot was used to assay the expression of protein levels. Here, we show that lidocaine could increase the survival rate of ALI mice and ameliorate the lung injury of ALI mice including reducing the edema, neutrophil infiltration, and pulmonary thrombosis formation and increasing blood flow velocity. Moreover, in vitro and in vivo, lidocaine could increase the expression of p-AMPK and SOCS3 and subsequently decrease the expression of p-ASK1, p-p38, TF, and the activity of MMP-2/9. Taken together, our study demonstrated that lidocaine could inhibit the TLR4/ASK1/TF pathway to alleviate ALI via activating AMPK-SOCS3 axis.
Collapse
|
10
|
Qian XP, Zhang XH, Sun LN, Xing WF, Wang Y, Sun SY, Ma MY, Cheng ZP, Wu ZD, Xing C, Chen BN, Wang YQ. Corosolic acid and its structural analogs: A systematic review of their biological activities and underlying mechanism of action. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153696. [PMID: 34456116 DOI: 10.1016/j.phymed.2021.153696] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The corosolic acid (CA), also known as plant insulin, is a pentacyclic triterpenoid extracted from plants such as Lagerstroemia speciosa. It has been shown to have anti-diabetic, anti-inflammatory and anti-tumor effects. Its structural analogs ursolic acid (UA), oleanolic acid (OA), maslinic acid (MA), asiatic acid (AA) and betulinic acid (BA) display similar individual pharmacological activities to those of CA. However, there is no systematic review documenting pharmacological activities of CA and its structural analogues. This study aims to fill this gap in literature. PURPOSE This systematic review aims to summarize the medical applications of CA and its analogues. METHODS A systematic review summarizes and compares the extraction techniques, pharmacokinetic parameters, and pharmacological effects of CA and its structural analogs. Hypoglycemic effect is one of the key inclusion criteria for searching Web of Science, PubMed, Embase and Cochrane databases up to October 2020 without language restrictions. 'corosolic acid', 'ursolic acid', 'oleanolic acid', 'maslinic acid', 'asiatic acid', 'betulinic acid', 'extraction', 'pharmacokinetic', 'pharmacological' were used to extract relevant literature. The PRISMA guidelines were followed. RESULTS At the end of the searching process, 140 articles were selected for the systematic review. Information of CA and five of its structural analogs including UA, OA, MA, AA and BA were included in this review. CA and its structural analogs are pentacyclic triterpenes extracted from plants and they have low solubilities in water due to their rigid scaffold and hydrophobic properties. The introduction of water-soluble groups such as sugar or amino groups could increase the solubility of CA and its structural analogs. Their biological activities and underlying mechanism of action are reviewed and compared. CONCLUSION CA and its structural analogs UA, OA, MA, AA and BA are demonstrated to show activities in lowering blood sugar, anti-inflammation and anti-tumor. Their oral absorption and bioavailability can be improved through structural modification and formulation design. CA and its structural analogs are promising natural product-based lead compounds for further development and mechanistic studies.
Collapse
Affiliation(s)
- Xu-Ping Qian
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China; Xuzhou Medical University, Xuzhou, China
| | - Xue-Hui Zhang
- Department of Pharmacy, Jiangsu Shengze Hospital, Nanjing Medical University, Suzhou, China
| | - Lu-Ning Sun
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China
| | - Wei-Fan Xing
- Nanjing Chenxiang Pharmaceutical Research Co. Ltd
| | - Yu Wang
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China
| | - Shi-Yu Sun
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China
| | - Meng-Yuan Ma
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China; Xuzhou Medical University, Xuzhou, China
| | - Zi-Ping Cheng
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China
| | - Zu-Dong Wu
- Nanjing Chenxiang Pharmaceutical Research Co. Ltd
| | - Chen Xing
- Nanjing Chenxiang Pharmaceutical Research Co. Ltd
| | - Bei-Ning Chen
- Department of Chemistry, University of Sheffield, Brookhill, Sheffield S3 7HF, United Kingdom.
| | - Yong-Qing Wang
- Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, Nanjing, China; Department of Pharmacy, Jiangsu Shengze Hospital, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
11
|
Wang YW, Wu YH, Zhang JZ, Tang JH, Fan RP, Li F, Yu BY, Kou JP, Zhang YY. Ruscogenin attenuates particulate matter-induced acute lung injury in mice via protecting pulmonary endothelial barrier and inhibiting TLR4 signaling pathway. Acta Pharmacol Sin 2021; 42:726-734. [PMID: 32855531 PMCID: PMC8114925 DOI: 10.1038/s41401-020-00502-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/31/2020] [Indexed: 12/15/2022] Open
Abstract
The inhalation of particulate matter (PM) is closely related to respiratory damage, including acute lung injury (ALI), characterized by inflammatory fluid edema and disturbed alveolar-capillary permeability. Ruscogenin (RUS), the main active ingredient in the traditional Chinese medicine Ophiopogonis japonicus, has been found to exhibit anti-inflammatory activity and rescue LPS-induced ALI. In this study, we investigated whether and how RUS exerted therapeutic effects on PM-induced ALI. RUS (0.1, 0.3, 1 mg·kg-1·d-1) was orally administered to mice prior to or after intratracheal instillation of PM suspension (50 mg/kg). We showed that RUS administration either prior to or after PM challenge significantly attenuated PM-induced pathological injury, lung edema, vascular leakage and VE-cadherin expression in lung tissue. RUS administration significantly decreased the levels of cytokines IL-6 and IL-1β, as well as the levels of NO and MPO in both bronchoalveolar lavage fluid (BALF) and serum. RUS administration dose-dependently suppressed the phosphorylation of NF-κB p65 and the expression of TLR4 and MyD88 in lung tissue. Furthermore, TLR4 knockout partly diminished PM-induced lung injury, and abolished the protective effects of RUS in PM-instilled mice. In conclusion, RUS effectively alleviates PM-induced ALI probably by inhibition of vascular leakage and TLR4/MyD88 signaling. TLR4 might be crucial for PM to initiate pulmonary lesion and for RUS to exert efficacy against PM-induced lung injury.
Collapse
Affiliation(s)
- Yu-Wei Wang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yun-Hao Wu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Zhi Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jia-Hui Tang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Rui-Ping Fan
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Fang Li
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bo-Yang Yu
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jun-Ping Kou
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Yuan-Yuan Zhang
- State Key Laboratory of Natural Products, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
12
|
Trushna T, Tripathi AK, Rana S, Tiwari RR. Nutraceuticals with anti-inflammatory and anti-oxidant properties as intervention for reducing the health effects of fine particulate matter: Potential and Prospects. Comb Chem High Throughput Screen 2021; 25:1639-1660. [PMID: 33845731 DOI: 10.2174/1386207324666210412121226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 11/22/2022]
Abstract
Air pollution, especially particulate matter pollution adversely affects human health. A growing pool of evidence has emerged which underscores the potential of individual-level nutritional interventions in attenuating the adverse health impact of exposure to PM2.5. Although controlling emission and reducing the overall levels of air pollution remains the ultimate objective globally, the sustainable achievement of such a target and thus consequent protection of human health will require a substantial amount of time and concerted efforts worldwide. In the meantime, smaller-scale individual-level interventions that can counter the inflammatory or oxidative stress effects triggered by exposure to particulate matter may be utilized to ameliorate the health effects of PM2.5 pollution. One such intervention is incorporation of nutraceuticals in the diet. Here, we present a review of the evidence generated from various in vitro, in vivo and human studies regarding the effects of different anti-inflammatory and antioxidant nutraceuticals in ameliorating the health effects of particulate matter air pollution. The studies discussed in this review suggest that these nutraceuticals when consumed as a part of the diet, or as additional supplementation, can potentially negate the cellular level adverse effects of exposure to particulate pollution. The potential benefits of adopting a non-pharmacological diet-based approach to air pollution-induced disease management have also been discussed. We argue that before a nutraceuticals-based approach can be used for widespread public adoption, further research, especially human clinical trials, is essential to confirm the beneficial action of relevant nutraceuticals and to explore the safe limits of human supplementation and the risk of side effects. Future research should focus on systematically translating bench-based knowledge regarding nutraceuticals gained from in-vitro and in-vivo studies into clinically usable nutritional guidelines.
Collapse
Affiliation(s)
- Tanwi Trushna
- Department of Environmental Health and Epidemiology, ICMR- National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Amit K Tripathi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Sindhuprava Rana
- Department of Bioinformatics, ICMR-National Institute for Research in Environmental Health, Bhopal- 462030. India
| | - Rajnarayan R Tiwari
- ICMR- National Institute for Research in Environmental Health (NIREH), Bhopal-462030, Madhya Pradesh. India
| |
Collapse
|
13
|
Wu Y, Xiao W, Pei C, Wang M, Wang X, Huang D, Wang F, Wang Z. Astragaloside IV alleviates PM2.5-induced lung injury in rats by modulating TLR4/MyD88/NF-κB signalling pathway. Int Immunopharmacol 2021; 91:107290. [PMID: 33383446 DOI: 10.1016/j.intimp.2020.107290] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/25/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Astragaloside IV (AS IV) is antioxidant and anti-inflammatory product, which is extracted from the Chinese herb Astragalus membranaceus. It is widely used in a variety of inflammatory diseases. The research was to explored the protective effects of AS IV against lung injury induced by particulate matter 2.5 (PM2.5) in vivo. SUBJECTS AND METHODS Thirty-five male Sprague-Dawley rats were randomly divided into five groups (n=7 per group). (1) Normal saline group (NS), (2) AS IV group (AS) (100 mg/kg), (3) PM2.5 group (PM2.5), (4) PM2.5 + AS IV group (ASL) (50 mg/kg), and (5) PM2.5 + AS IVgroup (ASH) (100 mg/kg). Rats were pre-treated with AS IV intraperitoneally (50 and 100 mg/kg/day) for three days. Then, PM2.5 (7.5 mg/kg) was given by intratracheal instillation to induce lung injury. Six hours after PM2.5 stimulation, the rats were euthanized. Bronchoalveolar lavage fluid (BALF) was collected for assay of cytokines. Lung tissue was collected for oxidative stress, histology, immunohistochemistry, transmission electron microscope, and western blot analyses. RESULTS AS IV alleviated PM2.5-induced lung injury by decreasing lung dry-wet ratio, reducing the level of interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α), and C-reactive protein (CRP) in BALF, and reduced oxidative stress response in lung tissue. Western blot results revealed that AS-IV regulated the expression of TLR4/MyD88/NF-κB pathway proteins in lung tissues. CONCLUSION AS IV mitigated PM2.5 induced lung injury by regulating the activity of TLR4/MyD88/NF-κB signalling pathway, reducing inflammatory and oxidative stress responses.
Collapse
Affiliation(s)
- Yongcan Wu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Wei Xiao
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Caixia Pei
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Mingjie Wang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Xiaomin Wang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Demei Huang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Fei Wang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China
| | - Zhenxing Wang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|