1
|
Luo M, Jin T, Fang Y, Chen F, Zhu L, Bai J, Ding J. Signaling Pathways Involved in Acute Pancreatitis. J Inflamm Res 2025; 18:2287-2303. [PMID: 40230438 PMCID: PMC11995411 DOI: 10.2147/jir.s485804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/25/2025] [Indexed: 04/16/2025] Open
Abstract
Acute pancreatitis (AP) is a common digestive emergency with high morbidity and mortality. Over the past decade, significant progress has been made in understanding the mechanisms of AP, including oxidative stress, disruptions in calcium homeostasis, endoplasmic reticulum stress, inflammatory responses, and various forms of cell death. This review provides an overview of the typical signaling pathways involved and proposes the latest clinical translation prospects. These strategies are important for the early management of AP, preventing multi-organ injury, and improving the overall prognosis of the disease.
Collapse
Affiliation(s)
- Mengchen Luo
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Ting Jin
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Yi Fang
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Feng Chen
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Lujian Zhu
- Department of Infectious Diseases, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, People’s Republic of China
| | - Jin Ding
- Department of Gastroenterology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People’s Republic of China
| |
Collapse
|
2
|
Tao Q, Zhang ZD, Qin Z, Liu XW, Li SH, Bai LX, Ge WB, Li JY, Yang YJ. Aspirin eugenol ester alleviates lipopolysaccharide-induced acute lung injury in rats while stabilizing serum metabolites levels. Front Immunol 2022; 13:939106. [PMID: 35967416 PMCID: PMC9372404 DOI: 10.3389/fimmu.2022.939106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Aspirin eugenol ester (AEE) was a novel drug compound with aspirin and eugenol esterified. AEE had various pharmacological activities, such as anti-inflammatory, antipyretic, analgesic, anti-oxidative stress and so on. In this study, it was aimed to investigate the effect of AEE on the acute lung injury (ALI) induced by lipopolysaccharide (LPS) in rats. In vitro experiments evaluated the protective effect of AEE on the LPS-induced A549 cells. The tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) were measured in the cell supernatant. The Wistar rats were randomly divided into five groups (n = 8): control group, model group (LPS group), LPS + AEE group (AEE, 54 mg·kg-1), LPS + AEE group (AEE, 108 mg·kg-1), LPS + AEE group (AEE, 216 mg·kg-1). The lung wet-to-dry weight (W/D) ratio and immune organ index were calculated. WBCs were counted in bronchoalveolar lavage fluid (BALF) and total protein concentration was measured. Hematoxylin-Eosin (HE) staining of lung tissue was performed. Glutathione (GSH), glutathione peroxidase (GPx), catalase (CAT), antioxidant superoxide dismutase (SOD), total antioxidant capacity (T-AOC), lactate dehydrogenase (LDH), C-reactive protein (CRP), myeloperoxidase (MPO), malondialdehyde (MDA), macrophage mobility inhibitory factor (MIF), TNF-α, IL-6, and IL-1β activity were measured. The metabolomic analysis of rat serum was performed by UPLC-QTOF-MS/MS. From the results, compared with LPS group, AEE improved histopathological changes, reduced MDA, CRP, MPO, MDA, and MIF production, decreased WBC count and total protein content in BALF, pro-inflammatory cytokine levels, immune organ index and lung wet-dry weight (W/D), increased antioxidant enzyme activity, in a dose-dependent manner. The results of serum metabolomic analysis showed that the LPS-induced ALI caused metabolic disorders and oxidative stress in rats, while AEE could ameliorate it to some extent. Therefore, AEE could alleviate LPS-induced ALI in rats by regulating abnormal inflammatory responses, slowing down oxidative stress, and modulating energy metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
3
|
Drug D, a Diosgenin Derive, Inhibits L-Arginine-Induced Acute Pancreatitis through Meditating GSDMD in the Endoplasmic Reticulum via the TXNIP/HIF-1α Pathway. Nutrients 2022; 14:nu14132591. [PMID: 35807771 PMCID: PMC9268286 DOI: 10.3390/nu14132591] [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: 05/13/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 12/13/2022] Open
Abstract
Acute pancreatitis (AP) is one of the most common causes of hospitalization for gastrointestinal diseases, with high morbidity and mortality. Endoplasmic reticulum stress (ERS) and Gasdermin D (GSDMD) mediate AP, but little is known about their mutual influence on AP. Diosgenin has excellent anti-inflammatory and antioxidant effects. This study investigated whether Diosgenin derivative D (Drug D) inhibits L-arginine-induced acute pancreatitis through meditating GSDMD in the endoplasmic reticulum (ER). Our studies were conducted in a mouse model of L-arginine-induced AP as well as in an in vitro model on mouse pancreatic acinar cells. The GSDMD accumulation in ER was found in this study, which caused ERS of acinar cells. GSDMD inhibitor Disulfiram (DSF) notably decreased the expression of GSDMD in ER and TXNIP/HIF-1α signaling. The molecular docking study indicated that there was a potential interaction between Drug D and GSDMD. Our results showed that Drug D significantly inhibited necrosis of acinar cells dose-dependently, and we also found that Drug D alleviated pancreatic necrosis and systemic inflammation by inhibiting the GSDMD accumulation in the ER of acinar cells via the TXNIP/HIF-1α pathway. Furthermore, the level of p-IRE1α (a marker of ERS) was also down-regulated by Drug D in a dose-dependent manner in AP. We also found that Drug D alleviated TXNIP up-regulation and oxidative stress in AP. Moreover, our results revealed that GSDMD-/- mitigated AP by inhibiting TXNIP/HIF-1α. Therefore, Drug D, which is extracted from Dioscorea zingiberensis, may inhibit L-arginine-induced AP by meditating GSDMD in the ER by the TXNIP /HIF-1α pathway.
Collapse
|
4
|
Prophylactic Penehyclidine Inhalation for Prevention of Postoperative Pulmonary Complications in High-risk Patients: A Double-blind Randomized Trial. Anesthesiology 2022; 136:551-566. [PMID: 35226725 DOI: 10.1097/aln.0000000000004159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Postoperative pulmonary complications are common. Aging and respiratory disease provoke airway hyperresponsiveness, high-risk surgery induces diaphragmatic dysfunction, and general anesthesia contributes to atelectasis and peripheral airway injury. This study therefore tested the hypothesis that inhalation of penehyclidine, a long-acting muscarinic antagonist, reduces the incidence of pulmonary complications in high-risk patients over the initial 30 postoperative days. METHODS This single-center double-blind trial enrolled 864 patients age over 50 yr who were scheduled for major upper-abdominal or noncardiac thoracic surgery lasting 2 h or more and who had an Assess Respiratory Risk in Surgical Patients in Catalonia score of 45 or higher. The patients were randomly assigned to placebo or prophylactic penehyclidine inhalation from the night before surgery through postoperative day 2 at 12-h intervals. The primary outcome was the incidence of a composite of pulmonary complications within 30 postoperative days, including respiratory infection, respiratory failure, pleural effusion, atelectasis, pneumothorax, bronchospasm, and aspiration pneumonitis. RESULTS A total of 826 patients (mean age, 64 yr; 63% male) were included in the intention-to-treat analysis. A composite of pulmonary complications was less common in patients assigned to penehyclidine (18.9% [79 of 417]) than those receiving the placebo (26.4% [108 of 409]; relative risk, 0.72; 95% CI, 0.56 to 0.93; P = 0.010; number needed to treat, 13). Bronchospasm was less common in penehyclidine than placebo patients: 1.4% (6 of 417) versus 4.4% (18 of 409; relative risk, 0.327; 95% CI, 0.131 to 0.82; P = 0.011). None of the other individual pulmonary complications differed significantly. Peak airway pressures greater than 40 cm H2O were also less common in patients given penehyclidine: 1.9% (8 of 432) versus 4.9% (21 of 432; relative risk, 0.381; 95% CI, 0.171 to 0.85; P = 0.014). The incidence of other adverse events, including dry mouth and delirium, that were potentially related to penehyclidine inhalation did not differ between the groups. CONCLUSIONS In high-risk patients having major upper-abdominal or noncardiac thoracic surgery, prophylactic penehyclidine inhalation reduced the incidence of pulmonary complications without provoking complications. EDITOR’S PERSPECTIVE
Collapse
|
5
|
Kitzerow O, Zucker IH, Lisco SJ, Wang HJ. Timeline of Multi-Organ Plasma Extravasation After Bleomycin-Induced Acute Lung Injury. Front Physiol 2022; 13:777072. [PMID: 35173628 PMCID: PMC8841715 DOI: 10.3389/fphys.2022.777072] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/10/2022] [Indexed: 12/30/2022] Open
Abstract
Acute lung injury (ALI) is characterized by the abrupt onset of clinically significant hypoxemia in the context of non-hydrostatic pulmonary edema. Acute lung injury is associated with cytokine release and plasma extravasation (PEx) that can cause pulmonary edema and subsequently acute respiratory distress syndrome (ARDS). Therefore, it is critical we understand the relationship between ALI and lung PEx. In addition, it is also important to assess PEx in the lungs and other organs post-ALI since ALI/ARDS often causes multi-organ failure. We hypothesized that ALI induces time-dependent lung PEx, which promotes extravasation in the heart, liver, kidney, spleen, pancreas, and gastrointestinal (GI) tract, in a time-dependent manner. To test our hypothesis, we administered bleomycin or saline via tracheal intubation in 8-week-old Sprague Dawley rats. At the terminal experiments, Evans Blue was injected (IV) through the femoral vein to allow for the visualization of PEx. Plasma extravasation of desired organs was evaluated at 3-, 7-, 14-, 21-, and 28-days after bleomycin or saline treatment by evaluating Evans Blue concentrations calorimetrically at fluorescence excitation wavelength of 620 nm (bandwidth 10 nm) and an emission wavelength of 680 nm (bandwidth 40 nm). Data show that ALI induces lung PEx beginning at day 3 and peaking between 7 and 21 days. Extravasation was also seen in all organs at varying degrees beginning at day 3 and peaking between days 7 and 14. Resolution appears to start after day 21 and continues past day 28. We conclude that ALI caused by bleomycin incites a time-dependent PEx of the lungs and multiple other organs.
Collapse
Affiliation(s)
- Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Irving H. Zucker
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Steven J. Lisco
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Han-Jun Wang
- Deptrtment of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Han-Jun Wang,
| |
Collapse
|
6
|
Yang K, Li B, Chen J. Knockdown of phosphoinositide-dependent kinase 1 (PDK1) inhibits fibrosis and inflammation in lipopolysaccharide-induced acute lung injury rat model by attenuating NF-κB/p65 pathway activation. ANNALS OF TRANSLATIONAL MEDICINE 2022; 9:1671. [PMID: 34988180 PMCID: PMC8667129 DOI: 10.21037/atm-21-5476] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/09/2021] [Indexed: 01/11/2023]
Abstract
Background Acute lung injury (ALI) is a common inflammatory disease of the lung. This study aimed to investigate the effect of 3-phosphoinositide-dependent kinase 1 (PDK1) interference on the levels of fibrosis and proinflammatory factors in lipopolysaccharide (LPS)-induced ALI and discuss the relevant mechanism. Methods An ALI model was established by intravenous injection of LPS treatment. A total of 24 Sprague-Dawley (SD) rats were randomly divided into 4 groups: sham group; ALI group; ALI + shRNA-NC group; and ALI + PDK1-shRNA group. Lung injury score, minute ventilation, lung volume, and airway resistance were used to evaluate lung function injury. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect PDK1 messenger RNA (mRNA) level. Western blot was performed to detect expression levels of PDK1, transforming growth factor-β (TGF-β), α-smooth muscle actin (α-SMA), toll-like receptor 4 (TLR4), p65, and myeloid differentiation primary response gene 88 (MyD88). The contents of interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) were detected by enzyme-linked immunosorbent assay (ELISA). The pathological changes and fibrosis of lung tissues were estimated by hematoxylin and eosin (H&E) and Masson staining. Results The results revealed that high lung injury score, low minute ventilation, low lung volume, and small airway resistance were present in the ALI group. Likewise, severe histopathological damage and fibrosis were apparent in the ALI group. Otherwise, contents of TNF-α, iNOS, IL-6, MCP-1, and levels of α-SMA, TGF-β, TLR4, phosphorylated (p)-p65, and MyD88 were enhanced in the ALI group. Interestingly, pathological changes and fibrosis were improved significantly in the ALI + PDK1-shRNA group. Besides, knockdown of PDK1 reduced lung injury score and enhanced minute ventilation, lung volume, and airway resistance. Moreover, knockdown of PDK1 decreased the contents of TNF-α, iNOS, IL-6, MCP-1, and levels of TGF-β, α-SMA, TLR4, p-p65, and MyD88. Conclusions Knockdown of PDK1 protects LPS-induced ALI via attenuating activation of the nuclear factor-κB (NF-κB)/p65 pathway.
Collapse
Affiliation(s)
- Keke Yang
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, China
| | - Boqian Li
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, China
| | - Jinghua Chen
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, China
| |
Collapse
|
7
|
Zhou L, Chen J, Mu G, Lu Z, Li W, Deng Y. Heparin-binding protein (HBP) worsens the severity of pancreatic necrosis via up-regulated M1 macrophages activation in acute pancreatitis mouse models. Bioengineered 2021; 12:11978-11986. [PMID: 34895060 PMCID: PMC8810113 DOI: 10.1080/21655979.2021.2011018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022] Open
Abstract
Acute pancreatitis (AP) is one of the most widespread clinical emergencies. Macrophages are the most common immune cells in AP pancreatic tissue and are closely associated with pancreatic necrosis and recovery. The level of heparin-binding protein (HBP) is closely linked to inflammation. In this study, we assessed the effect of HBP on AP tissue necrosis severity and whether HBP is associated with M1 macrophages in pancreatic necrosis. We observed the dynamic changes of HBP levels in the pancreas during acute inflammation in the caerulein-induced AP mice model. We used hematoxylin-eosin staining to evaluate pancreatic edema and necrosis, and to detect infiltration of macrophages by immunohistochemistry. Moreover, expressions of the maker and cytokines of macrophages, including inducible nitric oxide synthase (iNOS), and arginase 1 (Arg-1), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) mRNA, were detected by real-time polymerase-chain reaction (RT-PCR). High levels of HBP in the pancreas were detected at 48 h, and heparin inhibited HBP expression in AP pancreatic tissue. Inhibiting HBP expression by injecting heparin before AP can alleviate pancreatic necrosis and inhibit F4/80 labeled M1 macrophage infiltration and IL-6, TNF-α, and iNOS mRNA expression. Clodronate liposome (CLDL) intraperitoneally treated mice showed no change in pancreatic HBP levels, but pancreatic macrophage-specific antigen F4/80 and TNF-α, IL-1β, and IL-6 mRNA levels decreased after CLDL treatment. HBP is critical for pancreatic necrosis response in acute pancreatitis by increasing the infiltration of M1 macrophages and promoting the secretion of inflammatory factors, such as TNF-α, IL-6, IL-1β, which can be reduced by heparin.
Collapse
Affiliation(s)
- Liangliang Zhou
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Jianjun Chen
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Genhua Mu
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Zhongqian Lu
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Weiqin Li
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Yijun Deng
- Department of Intensive Care Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School/The First People’s Hospital of Yancheng, Yancheng, Jiangsu Province, China
| |
Collapse
|
8
|
Lin T, Song J, Pan X, Wan Y, Wu Z, Lv S, Mi L, Wang Y, Tian F. Downregulating Gasdermin D Reduces Severe Acute Pancreatitis Associated with Pyroptosis. Med Sci Monit 2021; 27:e927968. [PMID: 33582700 PMCID: PMC7891845 DOI: 10.12659/msm.927968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Intestinal injury plays a key role in the pathogenesis of severe acute pancreatitis (SAP). In this study, we investigated the protective function of downregulated Gasdermin D (GSDMD) in intestinal damage in a mouse model of severe acute pancreatitis (SAP). Material/Methods Twenty-four healthy male C57BL/6 mice were randomly divided into 4 groups – the NS group, the siRNA-NS group, the SAP group, and the siRNA-SAP group – with 6 mice in each group. SAP was induced in mice by intraperitoneal injection of caerulein and lipopolysaccharide. The pathological changes of pancreatic and the intestinal mucosa and the relative gene and protein expressions in each group were compared, and the levels of GSDMD and serum IL-1β and IL-18 were evaluated after induction of the SAP model. Results The mice in the SAP group were in more serious condition than those in the siRNA-SAP group, with various degrees of edema and hemorrhage in the intestinal tract. Under an optical microscope, the pathological changes of pancreatic tissue such as edema, inflammatory cell infiltration, and the damage of lobular structural were gradually increased in the SAP group and the siRNA-NS group. In addition, intestinal mucosal damage and intestinal villus breakage were found in the SAP group and the siRNA-NS group, and the latter was lighter than the former. Compared with the SAP group, the level of GSDMD protein expression in the siRNA-SAP group was lower, and the serum levels of IL-1β and IL-18 were higher in the SAP group and siRNA-SAP group (P<0.05). Immunohistochemical analysis showed the occludin and ZO-1 proteins in the NS group had a strong brown linear signal, while the brown-positive signals were weaker in the siRNA-SAP group and the SAP group. Conclusions Downregulating GSDMD protein can reduce pancreatitis associated with pyroptosis.
Collapse
Affiliation(s)
- Tianjiao Lin
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Jingyu Song
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Xinting Pan
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Youdong Wan
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Ziqian Wu
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Shaoyan Lv
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Liangyu Mi
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Yunyun Wang
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Fei Tian
- Emergency Intensive Care Unit, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
9
|
Liu Y, Xiang D, Zhang H, Yao H, Wang Y. Hypoxia-Inducible Factor-1: A Potential Target to Treat Acute Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8871476. [PMID: 33282113 PMCID: PMC7685819 DOI: 10.1155/2020/8871476] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is an acute hypoxic respiratory insufficiency caused by various intra- and extrapulmonary injury factors. Presently, excessive inflammation in the lung and the apoptosis of alveolar epithelial cells are considered to be the key factors in the pathogenesis of ALI. Hypoxia-inducible factor-1 (HIF-1) is an oxygen-dependent conversion activator that is closely related to the activity of reactive oxygen species (ROS). HIF-1 has been shown to play an important role in ALI and can be used as a potential therapeutic target for ALI. This manuscript will introduce the progress of HIF-1 in ALI and explore the feasibility of applying inhibitors of HIF-1 to ALI, which brings hope for the treatment of ALI.
Collapse
Affiliation(s)
- Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Hengcheng Zhang
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115 MA, USA
| | - Hanlin Yao
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan 430071, China
| |
Collapse
|
10
|
Effects of Bacterial Translocation and Autophagy on Acute Lung Injury Induced by Severe Acute Pancreatitis. Gastroenterol Res Pract 2020; 2020:8953453. [PMID: 32104173 PMCID: PMC7040400 DOI: 10.1155/2020/8953453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/22/2020] [Indexed: 12/27/2022] Open
Abstract
Aim To reveal the role of bacterial translocation (BT) and autophagy in severe acute pancreatitis-induced acute lung injury (SAP-ALI). Methods Rats were separated into a control (sham-operation) group (n = 10) and a SAP group (n = 10) and a SAP group ( Results Levels of TNF-α, IL-6, lipase, and amylase in the SAP group were significantly higher than those in the control group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group (P < 0.01). Histopathological score and W/D ratio of the lung in the SAP-BT(+) group were significantly higher than that in the SAP-BT(-) group ( Conclusions BT can aggravate SAP-ALI with the increasing oxidative stress level, which may be related to the decrease of autophagy level.
Collapse
|
11
|
Ye S, Yang X, Wang Q, Chen Q, Ma Y. Penehyclidine Hydrochloride Alleviates Lipopolysaccharide-Induced Acute Lung Injury by Ameliorating Apoptosis and Endoplasmic Reticulum Stress. J Surg Res 2019; 245:344-353. [PMID: 31425874 DOI: 10.1016/j.jss.2019.07.080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/16/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Penehyclidine hydrochloride (PHC), a novel anticholinergic reagent, has been shown to exert anti-endoplasmic reticulum stress (ERS), antioxidant, and antiinflammation functions in various rat models. However, the definite pathogenesis of lung defensive roles of PHC remains unclear. This study measured the functions of PHC on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in rats. METHODS In this research, the LPS-induced ALI model was assessed through the branchial injection of LPS for 24 h. Male Sprague-Dawley rats were randomly allocated into 5 groups: sham, LPS, LPS + PHC (0.5 mg/kg), LPS + PHC (1 mg/kg), and LPS + PHC (2.5 mg/kg). The concentrations of superoxide dismutase, malondialdehyde, myeloperoxidase, and glutathione peroxidase were measured by enzyme-linked immunosorbent assay and immunohistochemistry analysis. Western blotting, real-time PCR, and immunofluorescence analysis were used to determine the ERS-associated protein levels and mRNA expression. The protein levels of Bax, Bcl-2, caspase-3, and caspase-9 were used to measure lung tissue apoptosis. RESULTS The results revealed that PHC administration inhibited LPS-induced ALI as indicated by the loss in the ratio of injury production evaluated through hematoxylin-eosin staining, in particular the lung sample sections, compared with the LPS group. PHC administration inhibited LPS-induced lung myeloperoxidase and serum concentrations of malondialdehyde, superoxide dismutase, and glutathione peroxidase in rats. PHC administration repressed the LPS-activated ERS-correlated pathway and apoptosis-associated protein levels in rats. CONCLUSIONS In summary, our findings indicated that PHC has a defensive effect on LPS-induced ALI by inhibiting oxidative stress, attenuating PERK and ATF6 signals, and suppressing ERS-mediated apoptosis.
Collapse
Affiliation(s)
- Shaobing Ye
- Department of Anesthesiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue Yang
- Department of Anesthesiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiansong Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Chen
- Department of Anesthesiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongda Ma
- Department of Anesthesiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Silva-Vaz P, Abrantes AM, Castelo-Branco M, Gouveia A, Botelho MF, Tralhão JG. Murine Models of Acute Pancreatitis: A Critical Appraisal of Clinical Relevance. Int J Mol Sci 2019; 20:E2794. [PMID: 31181644 PMCID: PMC6600324 DOI: 10.3390/ijms20112794] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/15/2022] Open
Abstract
Acute pancreatitis (AP) is a severe disease associated with high morbidity and mortality. Clinical studies can provide some data concerning the etiology, pathophysiology, and outcomes of this disease. However, the study of early events and new targeted therapies cannot be performed on humans due to ethical reasons. Experimental murine models can be used in the understanding of the pancreatic inflammation, because they are able to closely mimic the main features of human AP, namely their histologic glandular changes and distant organ failure. These models continue to be important research tools for the reproduction of the etiological, environmental, and genetic factors associated with the pathogenesis of this inflammatory pathology and the exploration of novel therapeutic options. This review provides an overview of several murine models of AP. Furthermore, special focus is made on the most frequently carried out models, the protocols used, and their advantages and limitations. Finally, examples are provided of the use of these models to improve knowledge of the mechanisms involved in the pathogenesis, identify new biomarkers of severity, and develop new targeted therapies.
Collapse
Affiliation(s)
- Pedro Silva-Vaz
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal.
- General Surgery Department, Unidade Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Ana Margarida Abrantes
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Miguel Castelo-Branco
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), 6200-506 Covilhã, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - António Gouveia
- General Surgery Department, Unidade Local de Saúde de Castelo Branco, 6000-085 Castelo Branco, Portugal.
- Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Maria Filomena Botelho
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
| | - José Guilherme Tralhão
- Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
- Biophysics and Biomathematics Institute, IBILI-Faculty of Medicine of University of Coimbra, 3000-548 Coimbra, Portugal.
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal.
| |
Collapse
|
13
|
Kong Q, Wu X, Duan W, Zhan L, Song X. Penehyclidine hydrochloride exerts protective effects in rats with acute lung injury via the Fas/FasL signaling pathway. Exp Ther Med 2019; 17:3598-3606. [PMID: 30988742 PMCID: PMC6447765 DOI: 10.3892/etm.2019.7340] [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: 06/14/2018] [Accepted: 01/01/2019] [Indexed: 11/28/2022] Open
Abstract
Acute lung injury (ALI) is a critical syndrome that is associated with high morbidity and mortality rates. The activation of the Fas/Fas ligand (FasL) signaling pathway may be an important pathophysiological mechanism during ALI development. Penehyclidine hydrochloride (PHC) has been revealed to exhibit anti-apoptotic properties and may attenuate the observed systemic inflammatory response. The present study was performed to elucidate the molecular mechanism of PHC in the regulation of the Fas/FasL signaling pathway in rats with ALI. An ALI rat model was constructed by inducing blunt chest trauma and hemorrhagic shock (T/HS), with PHC administration prior to or following T/HS. At 6 h following T/HS, blood samples and lung tissues were collected. Western blotting, arterial blood gas analysis, ELISA, hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and biochemical indicator analysis were performed to determine the degree of lung injury and the key signaling pathways associated with lung damage. The results indicated that the administration of PHC following T/HS effectively attenuates lung injury by improving pulmonary oxygenation, decreasing histopathological damage, decreasing polymorphonuclear neutrophil count and decreasing Fas, FasL, caspase-8, caspase-3, tumor necrosis factor-α, interleukin (IL)-6 and IL-1β expression. The results indicated that PHC exhibits anti-apoptotic functions and exerts protective effects in ALI rats induced by T/HS, which may be attributed to the inhibition of the Fas/FasL signaling pathway.
Collapse
Affiliation(s)
- Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weina Duan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Liying Zhan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuemin Song
- Department of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
14
|
Xie P, Zheng Z, Jiang L, Wu S. Penehyclidine effects the angiogenic potential of pulmonary microvascular endothelial cells. Pulm Pharmacol Ther 2019; 55:5-16. [PMID: 30641132 DOI: 10.1016/j.pupt.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/25/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
Abstract
The present study sought to determine the pharmacological effects of penehyclidine, an anticholinergic agent, on the angiogenic capacity of pulmonary microvascular endothelial cells (PMVECs). In vitro Matrigel network formation assay, cell proliferation assay, cell-matrix adhesion assay, and wound-healing assay were performed in PMVECs with or without exposure to penehyclidine or, in some cases, glycopyrrolate or acetylcholine, over a concentration range. In addition, the phosphorylation state of Akt and ERK, as well as the endogenous level of mTOR and RICTOR were examined in PMVECs by Western blot following the cells exposure to penehyclidine or, for some proteins, glycopyrrolate or acetylcholine. Finally, Western blot for Akt phosphorylation and in vitro Matrigel network formation assay were performed in PMVECs following their exposure to penehyclidine with or without phosphoinositide 3-kinase (PI3K) inhibitor LY294002 or mTOR inhibitor torin-1. We found that, in PMVECs, penehyclidine affected the network formation and cell migration, but not proliferation or cell-matrix adhesion, in a concentration-specific manner, i.e., penehyclidine increased the network formation and cell migration at lower concentrations but increased these processes at higher concentrations. Coincidentally, we observed that penehyclidine concentration-specifically affected the phosphorylation state of Akt in PMVECs, i.e., increased Akt phosphorylation at lower concentrations and decreased it at higher concentrations. In contrast, glycopyrrolate was found straightly to decrease network formation and Akt phosphorylation in a concentration-dependent manner. Further, we demonstrated that PI3K or mTOR blockade abolished both the enhanced network formation and the increased Akt phosphorylation by penehyclidine. Hence, penehyclidine may differentially alter the angiogenic capacity of PMVECs through affecting the Akt signaling pathway downstream of PI3K and mTOR. Findings from this study suggest a unique pharmacological feature of penehyclidine, which may imply its clinical and therapeutic value in modulating angiogenesis.
Collapse
Affiliation(s)
- Peilin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhen Zheng
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lihua Jiang
- Department of Anesthesiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|