1
|
Li Y, Zhao B, Peng J, Tang H, Wang S, Peng S, Ye F, Wang J, Ouyang K, Li J, Cai M, Chen Y. Inhibition of NF-κB signaling unveils novel strategies to overcome drug resistance in cancers. Drug Resist Updat 2024; 73:101042. [PMID: 38219532 DOI: 10.1016/j.drup.2023.101042] [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: 11/11/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/16/2024]
Abstract
Drug resistance in cancer remains a major challenge in oncology, impeding the effectiveness of various treatment modalities. The nuclear factor-kappa B (NF-κB) signaling pathway has emerged as a critical player in the development of drug resistance in cancer cells. This comprehensive review explores the intricate relationship between NF-κB and drug resistance in cancer. We delve into the molecular mechanisms through which NF-κB activation contributes to resistance against chemotherapeutic agents, targeted therapies, and immunotherapies. Additionally, we discuss potential strategies to overcome this resistance by targeting NF-κB signaling, such as small molecule inhibitors and combination therapies. Understanding the multifaceted interactions between NF-κB and drug resistance is crucial for the development of more effective cancer treatment strategies. By dissecting the complex signaling network of NF-κB, we hope to shed light on novel therapeutic approaches that can enhance treatment outcomes, ultimately improving the prognosis for cancer patients. This review aims to provide a comprehensive overview of the current state of knowledge on NF-κB and its role in drug resistance, offering insights that may guide future research and therapeutic interventions in the fight against cancer.
Collapse
Affiliation(s)
- Yuanfang Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer,Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Baiwei Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer,Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Juzheng Peng
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer,Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Sicheng Wang
- School of Medicine, Sun Yat-sen University, China
| | - Sicheng Peng
- School of Medicine, Sun Yat-sen University, China
| | - Feng Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer,Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Junye Wang
- School of Medicine, Sun Yat-sen University, China
| | - Kai Ouyang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jianjun Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Manbo Cai
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer,Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
2
|
Li H, Gao Y, Lin Y. Progress in molecular mechanisms of coronary microvascular dysfunction. Microcirculation 2023; 30:e12827. [PMID: 37608689 DOI: 10.1111/micc.12827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/23/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023]
Abstract
Coronary microvascular dysfunction is a high-risk factor for many cardiovascular events. However, because of multiple risk factors and limited understanding about its underlying pathophysiological mechanisms, it was easily misdiagnosed. Therefore, its clinical diagnosis and treatment were greatly restricted. Coronary microcirculation refers to microvessels that play an important role in the physiological regulation of myocardial perfusion and regulating blood flow distribution, fulfilling myocardial metabolic needs and moderating peripheral vascular resistance. In coronary microvascular dysfunction, vascular endothelial celldamage is a critical link. The main feature of early coronary microvascular dysfunction is the impairment of endothelial cell proliferation, adhesion, migration, apoptosis, and secretion. Moreover, coronary microvascular dysfunction risk factors include hyperglycemia, lipid metabolism disorders, ischemia-reperfusion injury, aging, and hypertension, similar to coronary atherosclerosis. There are various mechanisms by which these risk factors harm endothelial function and cause microcirculatory disturbances. Therefore, we reviewed coronary microvascular dysfunction's risk factors and pathogenesis in this article.
Collapse
Affiliation(s)
- Hao Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
3
|
Li L, Liu S, Tan J, Wei L, Wu D, Gao S, Weng Y, Chen J. Recent advance in treatment of atherosclerosis: Key targets and plaque-positioned delivery strategies. J Tissue Eng 2022; 13:20417314221088509. [PMID: 35356091 PMCID: PMC8958685 DOI: 10.1177/20417314221088509] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory disease of vascular wall, is a progressive pathophysiological process with lipids oxidation/depositing initiation and innate/adaptive immune responses. The coordination of multi systems covering oxidative stress, dysfunctional endothelium, diseased lipid uptake, cell apoptosis, thrombotic and pro-inflammatory responding as well as switched SMCs contributes to plaque growth. In this circumstance, inevitably, targeting these processes is considered to be effective for treating atherosclerosis. Arriving, retention and working of payload candidates mediated by targets in lesion direct ultimate therapeutic outcomes. Accumulating a series of scientific studies and clinical practice in the past decades, lesion homing delivery strategies including stent/balloon/nanoparticle-based transportation worked as the potent promotor to ensure a therapeutic effect. The objective of this review is to achieve a very brief summary about the effective therapeutic methods cooperating specifical targets and positioning-delivery strategies in atherosclerosis for better outcomes.
Collapse
Affiliation(s)
- Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Lai Wei
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Dimeng Wu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Shuai Gao
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| |
Collapse
|
4
|
Modified Yuejuwan Inhibited Cholesterol Accumulation and Inflammation in THP-1 Macrophage-Derived Foam Cells by Inhibiting the Activity of the TRIM37/TRAF2/NF- κB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6400517. [PMID: 35310029 PMCID: PMC8930229 DOI: 10.1155/2022/6400517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/25/2022] [Indexed: 01/25/2023]
Abstract
Background This study aimed to explore the function of modified Yuejuwan (MYJ) on THP-1 macrophage-derived foam cells. Methods First, human THP-cells were obtained, and then, grouping was made to the following: control group, foaming group, foaming group +0.2 mg/mL Jiawei Yueju pill, foaming group +0.5 mg/mL Jiawei Yueju pill, and foaming group +1 mg/mL Jiawei Yueju pill. An Oil Red O staining assay was used to examine the uptake of oxidatively modified low-density lipoprotein (oxLDL). The secretion of interleukin (IL)-1β and tumor necrosis factor (TNF)-α were determined using an enzyme-linked immunosorbent assay (ELISA). Real-time quantitative PCR (qRT-PCR) and Western blot were used to quantify genes and proteins expression levels. Results Our results indicated that MYJ inhibited the accumulation of total cholesterol (TC), free cholesterol (FC), and cholesteryl ester (CE) in foam cells. Moreover, the secretion of IL-1β and TNF-α also downregulated in foam cells after treatment of MYJ. Furthermore, we found that tripartite motif-containing 37 (TRIM37) was significantly upregulated in foam cells. Knockdown of TRIM37 promoted cholesterol efflux and presented an anti-inflammation effect in foam cells. Furthermore, TRIM37 positively mediated the translocation of NF-κB to nuclear. It negatively regulated its ubiquitination in foam cells after interacting with TRAF2. Importantly, MYJ profoundly suppressed the function of TRIM37 in foam cells and functioned as a TRIM37 inhibitor. Conclusions This study demonstrated that MYJ might alleviate oxLDL-induced foam cell formation by inhibiting the TRIM37/TRAF2/NF-κB pathway activity. MYJ was a potential agent in preventing atherosclerosis and indicated its potential signaling pathway in foam cells.
Collapse
|
5
|
Liu J, Sun Q, Sun M, Lin L, Ren X, Li T, Xu Q, Sun Z, Duan J. Melatonin alleviates PM 2.5-triggered macrophage M1 polarization and atherosclerosis via regulating NOX2-mediated oxidative stress homeostasis. Free Radic Biol Med 2022; 181:166-179. [PMID: 35149217 DOI: 10.1016/j.freeradbiomed.2022.02.005] [Citation(s) in RCA: 3] [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: 12/18/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/20/2022]
Abstract
It is reported that oxidative stress homeostasis was involved in PM2.5-induced foam cell formation and progression of atherosclerosis, but the exact molecular mechanism is still unclear. Melatonin is an effective antioxidant that could reverse the cardiopulmonary injury. The main purpose of this study is to investigate the latent mechanism of PM2.5-triggered atherosclerosis development and the protective role of melatonin administration. Vascular Doppler ultrasound showed that PM2.5 exposure reduced aortic elasticity in ApoE-/- mice. Meanwhile, blood biochemical and pathological analysis demonstrated that PM2.5 exposure caused dyslipidemia, elicited oxidative damage of aorta and was accompanied by an increase in atherosclerotic plaque area; while the melatonin administration could effectively alleviate PM2.5-induced macrophage M1 polarization and atherosclerosis in mice. Further investigation verified that NADPH oxidase 2 (NOX2) and mitochondria are two prominent sources of PM2.5-induced ROS production in vascular macrophages. Whereas, the combined use of two ROS-specific inhibitors and adopted with melatonin markedly rescued PM2.5-triggered macrophage M1 polarization and foam cell formation by inhibiting NOX2-mediated crosstalk of Keap1/Nrf2/NF-κB and TLR4/TRAF6/NF-κB signaling pathways. Our results demonstrated that NOX2-mediated oxidative stress homeostasis is critical for PM2.5-induced atherosclerosis and melatonin might be a potential treatment for air pollution-related cardiovascular diseases.
Collapse
Affiliation(s)
- Jiangyan Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Mengqi Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Lisen Lin
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qing Xu
- Core Facilities for Electrophysiology, Core Facilities Center, Capital Medical University, Beijing, 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
6
|
Tian H, Ding M, Guo Y, Zhu Z, Yu Y, Tian Y, Li K, Sun G, Jiang R, Han R, Yan F, Kang X. WITHDRAWN: Effect of HSPA8 on the proliferation, apoptosis and immune function of chicken macrophages. Int J Biochem Cell Biol 2022:106186. [PMID: 35217190 DOI: 10.1016/j.biocel.2022.106186] [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: 02/10/2022] [Accepted: 02/21/2022] [Indexed: 11/19/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal
Collapse
Affiliation(s)
- Huihui Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Mengxia Ding
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yujie Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhaoyan Zhu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yange Yu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Kui Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Guirong Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruirui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Ruili Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China
| | - Fengbin Yan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| | - Xiangtao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou 450046, China.
| |
Collapse
|
7
|
Chi X, Xiang D, Sha Y, Liang S, Wang C. Inhibition of human sperm function by an antibody against apolipoprotein A1: A protein located in human spermatozoa. Andrologia 2022; 54:e14365. [DOI: 10.1111/and.14365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/12/2021] [Accepted: 12/22/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Xiuping Chi
- Department of Laboratory Medicine the First Medical Centre, Chinese PLA General Hospita Beijing China
| | - Daijun Xiang
- Department of Laboratory Medicine the First Medical Centre, Chinese PLA General Hospita Beijing China
| | - Yingjiao Sha
- Department of Laboratory Medicine the First Medical Centre, Chinese PLA General Hospita Beijing China
| | - Shuang Liang
- Department of Medical Laboratory Maternal and Child Health Hospital Tangshan China
| | - Chengbin Wang
- Department of Laboratory Medicine the First Medical Centre, Chinese PLA General Hospita Beijing China
| |
Collapse
|
8
|
Fasolo F, Jin H, Winski G, Chernogubova E, Pauli J, Winter H, Li DY, Glukha N, Bauer S, Metschl S, Wu Z, Koschinsky ML, Reilly M, Pelisek J, Kempf W, Eckstein HH, Soehnlein O, Matic L, Hedin U, Bäcklund A, Bergmark C, Paloschi V, Maegdefessel L. Long Noncoding RNA MIAT Controls Advanced Atherosclerotic Lesion Formation and Plaque Destabilization. Circulation 2021; 144:1567-1583. [PMID: 34647815 PMCID: PMC8570347 DOI: 10.1161/circulationaha.120.052023] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Supplemental Digital Content is available in the text. Long noncoding RNAs (lncRNAs) are important regulators of biological processes involved in vascular tissue homeostasis and disease development. The present study assessed the functional contribution of the lncRNA myocardial infarction-associated transcript (MIAT) to atherosclerosis and carotid artery disease.
Collapse
Affiliation(s)
- Francesca Fasolo
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Hong Jin
- Department of Medicine (H.J., G.W., E.C., A.B.), Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery (H.J., L. Matic, U.H., C.B., L. Maegdefessel), Karolinska Institutet, Stockholm, Sweden
| | - Greg Winski
- Department of Medicine (H.J., G.W., E.C., A.B.), Karolinska Institutet, Stockholm, Sweden
| | - Ekaterina Chernogubova
- Department of Medicine (H.J., G.W., E.C., A.B.), Karolinska Institutet, Stockholm, Sweden
| | - Jessica Pauli
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Hanna Winter
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Daniel Y Li
- Department of Cardiology, Columbia University Medical Center, New York, NY (D.Y.L., M.R.)
| | - Nadiya Glukha
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Sabine Bauer
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Susanne Metschl
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Zhiyuan Wu
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | | | - Muredach Reilly
- Department of Cardiology, Columbia University Medical Center, New York, NY (D.Y.L., M.R.)
| | - Jaroslav Pelisek
- Department of Vascular Surgery, University Hospital Zurich, Switzerland (J. Pelisek)
| | - Wolfgang Kempf
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Oliver Soehnlein
- Department of Experimental Pathology, Westphalian Wilhelms University, Munster, Germany (O.S.).,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (O.S.).,Institute for Cardiovascular Prevention, Ludwig Maximilian University of Munich, Germany (O.S.)
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery (H.J., L. Matic, U.H., C.B., L. Maegdefessel), Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery (H.J., L. Matic, U.H., C.B., L. Maegdefessel), Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Bäcklund
- Department of Medicine (H.J., G.W., E.C., A.B.), Karolinska Institutet, Stockholm, Sweden
| | - Claes Bergmark
- Department of Molecular Medicine and Surgery (H.J., L. Matic, U.H., C.B., L. Maegdefessel), Karolinska Institutet, Stockholm, Sweden
| | - Valentina Paloschi
- German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel)
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany (F.F., J. Pauli, H.W., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,German Center for Cardiovascular Research (DZHK), Berlin, Germany; partner site Munich Heart Alliance (F.F., J. Pauli, H.W., F.F., N.G., S.B., S.M., Z.W., W.K., H.-H.E., V.P., L. Maegdefessel).,Department of Molecular Medicine and Surgery (H.J., L. Matic, U.H., C.B., L. Maegdefessel), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Tian H, Zhang Z, Han X, Pan T, Tao G, Jiao P, Zhai L, Yang L, Wang X, Yao Y, Qin S, Yao S. D4F alleviates the C/EBP homologous protein-mediated apoptosis in glycated high-density lipoprotein-treated macrophages by facilitating autophagy. Exp Biol Med (Maywood) 2021; 246:2595-2609. [PMID: 34525858 DOI: 10.1177/15353702211045323] [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: 11/15/2022] Open
Abstract
The present study aimed to investigate the role of D4F, an apolipoprotein A-I mimetic peptide, in macrophage apoptosis induced by the glycated high-density lipoprotein (gly-HDL)-induced endoplasmic reticulum (ER) stress C/EBP homologous protein (CHOP) pathway, and unravel the regulatory role of autophagy in this process. Our results revealed that except for suppressing the accumulation of lipids within RAW264.7 macrophages caused by gly-HDL, D4F inhibited gly-HDL-induced decrease in the cell viability and increase in lactate dehydrogenase leakage and cell apoptosis, which were similar to 4-phenylbutyric acid (PBA, an ER stress inhibitor). Besides, similar to PBA, D4F inhibited gly-HDL-induced ER stress response activation evaluated through the decreased PERK and eIF2α phosphorylation, together with reduced ATF6 nuclear translocation as well as the downregulation of GRP78 and CHOP. Interestingly, D4F facilitated gly-HDL-triggered activation of autophagy, measured as elevated levels of beclin-1, LC3-II, and ATG5 expressions in macrophages. Furthermore, the inhibition effect of D4F on gly-HDL-induced ER stress-CHOP-induced apoptosis of macrophages was restrained after beclin-1 siRNA and 3-methyladenine (3-MA, an inhibitor of autophagy) treatments, while this effect was further reinforced after rapamycin (Rapa, an inducer of autophagy) treatment. Furthermore, administering D4F or Rapa to T2DM mice upregulated LC3-II and attenuated CHOP expression, cell apoptosis, and atherosclerotic lesions. However, the opposite results were obtained when 3-MA was administered to these mice. These results support that D4F effectively protects macrophages against gly-HDL-induced ER stress-CHOP-mediated apoptosis by promoting autophagy.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhaoqiang Zhang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Xiaoyan Han
- College of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Tianqi Pan
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Geru Tao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Lei Zhai
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Libo Yang
- Department of Endocrinology, Central Hospital of Taian, Taian 271000, China
| | - Xiaoxu Wang
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Yilin Yao
- College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.,College of Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| |
Collapse
|
10
|
Kalantar K, Manzano-Román R, Ghani E, Mansouri R, Hatam G, Nguewa P, Rashidi S. Leishmanial apolipoprotein A-I expression: a possible strategy used by the parasite to evade the host's immune response. Future Microbiol 2021; 16:607-613. [PMID: 33998267 DOI: 10.2217/fmb-2020-0303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Apolipoprotein A-I (apo A-I) represents the main component of the Trypanosome lytic factor (TLF) which contributes to the host innate immunity against Trypanosoma and Leishmania. These parasites use complex and multiple strategies such as molecular mimicry to evade or subvert the host immune system. Previous studies have highlighted the adaptation mechanisms of TLF-resistant Trypanosoma species. These data might support the hypothesis that Leishmania parasites (amastigote forms in macrophages) might express apo A-I to bypass and escape from TLF action as a component of the host innate immune responses. The anti-inflammatory property of apo A-I is another mechanism that supports our idea that apo A-I may play a role in Leishmania parasites allowing them to bypass the host innate immune system.
Collapse
Affiliation(s)
- Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raúl Manzano-Román
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), Salamanca, 37007, Spain
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences & Health Services, Yazd, Iran
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- Department of Microbiology & Parasitology, University of Navarra, ISTUN Instituto de Salud Tropical, IdiSNA (Navarra Institute for Health Research), c/ Irunlarrea 1, Pamplona, 31008, Spain
| | - Sajad Rashidi
- Department of Parasitology & Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
11
|
Liu Z, Zhu H, Ma Y, Tang Z, Zhao N, Wang Y, Pan S. AGEs exacerbates coronary microvascular dysfunction in NoCAD by activating endoplasmic reticulum stress-mediated PERK signaling pathway. Metabolism 2021; 117:154710. [PMID: 33485865 DOI: 10.1016/j.metabol.2021.154710] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/18/2020] [Accepted: 01/20/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The current study was aimed to investigate the involvement of endoplasmic reticulum stress (ERS)-mediated protein kinase R-like endoplasmic reticulum kinase (PERK) signaling in advanced glycation end products (AGEs)-exacerbated coronary microvascular dysfunctions (CMD) in non-obstructive coronary artery disease (NoCAD). METHODS AND MATERIALS ob/ob-/- mice were used as NoCAD animal model which were exposed to AGEs by intraperitoneal injections. Animal CMD was evaluated by coronary flow velocity reserve (CFVR). A viral vector carrying perk-siRNA was used to silence PERK in vivo and in vitro studies. Cell apoptosis was detected by TUNEL. Immunofluorescent staining was used to assess CD42c-positive cell number in cardiac sections and NFATc4 translocation in CMECs. Real-time PCR and Western blotting were used to evaluate the gene expression levels. Cytokine and AGEs concentrations were determined by ELISA. Enzymatic activity of CaN was measured by a colorimetric method. A registered cross sectional study consisted of 77 patients diagnosed as NoCAD was used to analyze the association between diabetes and CMD which was measured by index of microvascular resistance (IMR) with a pressure wire system. RESULTS Significant CMD was found in NoCAD mice compared with healthy control. AGEs exposure exacerbated CMD in NoCAD animals which was improved by PERK silencing. Phosphorylation of PERK, nuclear translocation of nuclear factor of activated T-cells (NFAT)c4, enzymatic activity of calcineurin (CaN), expression levels of Fas/FasL, production of interleukin (IL)6, tumor necrosis factor (TNF)α, cyclooxygenase (COX)2, thromboxane B (TXB)2 as well as apoptosis were suppressed by PERK silencing in cardiac microcirculation endothelial cells (CMECs) isolated from AGEs-exposed NoCAD mice and AGEs-treated primary CMECs. PERK silencing also reduced CD42c-postive cells number in cardiac tissue from AGEs-exposed NoCAD mice. CONCLUSION Diabetes was associated with CMD in NoCAD. AGEs fostered in diabetes exacerbated CMD by activating ERS-mediated PERK/CaN/NFATc4 signaling in CMECs. IMR values increased significantly in NoCAD patients complicated with diabetes, which were significantly and positively correlated with serum AGEs concentrations.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Haitao Zhu
- Department of Pediatrics, Northwest Women's and Children's Hospital, Xi'an, Shaanxi Province 710000, China
| | - Yanpeng Ma
- Department of Cardiology, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Zhiguo Tang
- Department of Cardiology, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Na Zhao
- Department of Cardiology, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China
| | - Yuan Wang
- Department of Preventive Medicine, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China.
| | - Shuo Pan
- Department of Cardiology, Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, Shaanxi Province 710072, China.
| |
Collapse
|
12
|
Paul S, Gangwar A, Bhargava K, Ahmad Y. D4F prophylaxis enables redox and energy homeostasis while preventing inflammation during hypoxia exposure. Biomed Pharmacother 2021; 133:111083. [PMID: 33378979 DOI: 10.1016/j.biopha.2020.111083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/18/2020] [Accepted: 11/28/2020] [Indexed: 02/02/2023] Open
Abstract
Apo-A1 is correlated with conditions like hyperlipidemia, cardiovascular diseases, high altitude pulmonary edema and etc. where hypoxia constitutes an important facet.Hypoxia causes oxidative stress, vaso-destructive and inflammatory outcomes.Apo-A1 is reported to have vasoprotective, anti-oxidative, anti-apoptotic, and anti-inflammatory effects. However, effects of Apo-A1 augmentation during hypoxia exposure are unknown.In this study, we investigated the effects of exogenously supplementing Apo-A1-mimetic peptide on SD rats during hypoxia exposure. For easing the processes of delivery, absorption and bio-availability, Apo-A1 mimetic peptide D4F was used. The rats were given 10 mg/kg BW dose (i.p.) of D4F for 7 days and then exposed to hypoxia. D4F was observed to attenuate both oxidative stress and inflammation during hypoxic exposure. D4F improved energy homeostasis during hypoxic exposure. D4F did not affect HIF-1a levels during hypoxia but increased MnSOD levels while decreasing CRP and Apo-B levels. D4F showed promise as a prophylactic against hypoxia exposure.
Collapse
Affiliation(s)
- Subhojit Paul
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Anamika Gangwar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Kalpana Bhargava
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India
| | - Yasmin Ahmad
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi, 110054, India.
| |
Collapse
|
13
|
Oehler B, Kloka J, Mohammadi M, Ben-Kraiem A, Rittner HL. D-4F, an ApoA-I mimetic peptide ameliorating TRPA1-mediated nocifensive behaviour in a model of neurogenic inflammation. Mol Pain 2020; 16:1744806920903848. [PMID: 31996074 PMCID: PMC6993174 DOI: 10.1177/1744806920903848] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background High doses of capsaicin are recommended for the treatment of neuropathic pain. However, low doses evoke mechanical hypersensitivity. Activation of the capsaicin chemosensor transient receptor potential vanilloid 1 (TRPV1) induces neurogenic inflammation. In addition to the release of pro-inflammatory mediators, reactive oxygen species are produced. These highly reactive molecules generate oxidised phospholipids and 4-hydroxynonenal (4-HNE) which then directly activate TRP ankyrin 1 (TRPA1). The apolipoprotein A-I mimetic peptide D-4F neutralises oxidised phospholipids. Here, we asked whether D-4F ameliorates neurogenic hypersensitivity in rodents by targeting reactive oxygen species and 4-HNE in the capsaicin-evoked pain model. Results Co-application of D-4F ameliorated capsaicin-induced mechanical hypersensitivity and allodynia as well as persistent heat hypersensitivity measured by Randell–Selitto, von Frey and Hargreaves test, respectively. In addition, mechanical hypersensitivity was blocked after co-injection of D-4F with the reactive oxygen species analogue H2O2 or 4-HNE. In vitro studies on dorsal root ganglion neurons and stably transfected cell lines revealed a TRPA1-dependent inhibition of the calcium influx when agonists were pre-incubated with D-4F. The capsaicin-induced calcium influx in TRPV1-expressing cell lines and dorsal root ganglion neurons sustained in the presence of D-4F. Conclusions D-4F is a promising compound to ameliorate TRPA1-dependent hypersensitivity during neurogenic inflammation.
Collapse
Affiliation(s)
- Beatrice Oehler
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Jan Kloka
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Department of Anaesthesiology, University Hospital of Frankfurt, Frankfurt, Germany
| | - Milad Mohammadi
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany.,Department of Anaesthesiology, University Hospital of Cologne, Cologne, Germany
| | - Adel Ben-Kraiem
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L Rittner
- Department of Anaesthesiology, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
Lambrou GI, Hatziagapiou K, Vlahopoulos S. Inflammation and tissue homeostasis: the NF-κB system in physiology and malignant progression. Mol Biol Rep 2020; 47:4047-4063. [PMID: 32239468 DOI: 10.1007/s11033-020-05410-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Disruption of tissue function activates cellular stress which triggers a number of mechanisms that protect the tissue from further damage. These mechanisms involve a number of homeostatic modules, which are regulated at the level of gene expression by the transactivator NF-κB. This transcription factor shifts between activation and repression of discrete, cell-dependent gene expression clusters. Some of its target genes provide feedback to NF-κB itself, thereby strengthening the inflammatory response of the tissue and later terminating inflammation to facilitate restoration of tissue homeostasis. Disruption of key feedback modules for NF-κB in certain cell types facilitates the survival of clones with genomic aberrations, and protects them from being recognized and eliminated by the immune system, to enable thereby carcinogenesis.
Collapse
Affiliation(s)
- George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece.
| |
Collapse
|
15
|
Antitumor activity of methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide against leukemia cell lines via mitotic arrest and apoptotic pathways. Biochim Biophys Acta Gen Subj 2019; 1863:1332-1342. [PMID: 31170497 DOI: 10.1016/j.bbagen.2019.05.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 01/07/2023]
Abstract
In a previous study, we described a series of 28 aryl- and alkyl-substituted isothiouronium salts with antitumor activity and selectivity toward a leukemia cell line. Among the synthesized compounds, methyl (Z)-2-(isothioureidomethyl)-2-pentenoate hydrobromide (IS-MF08) showed conspicuous activity. In the present study, we investigated the mechanism of action of IS-MF08. Our results showed that its mechanism most likely is related with the membrane receptor Fas and subsequent activation of the extrinsic cell death pathway, triggered by a decrease in the levels of the anti-apoptotic protein Bcl-2 and caspase-8 and -3 cascade activation, causing DNA damage and mitotic arrest. IS-MF08 also caused an increase in intracellular ROS, endoplasmic reticulum (ER) stress, and mitochondrial membrane permeabilization, resulting in organelle degradation as an attempt to reestablish cell homeostasis. Furthermore, cells exposed to IS-MF08 combined to an autophagy inhibitor were less susceptible to compound's cytotoxicity, suggesting that autophagy makes part of its mechanism of action. These data support the hypothesis that IS-MF08 acts by the apoptosis extrinsic pathway and possibly by autophagy as mechanisms of cell death.
Collapse
|
16
|
Martinet W, Coornaert I, Puylaert P, De Meyer GRY. Macrophage Death as a Pharmacological Target in Atherosclerosis. Front Pharmacol 2019; 10:306. [PMID: 31019462 PMCID: PMC6458279 DOI: 10.3389/fphar.2019.00306] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual build-up of plaques within the vessel wall of middle-sized and large arteries. Over the past decades, treatment of atherosclerosis mainly focused on lowering lipid levels, which can be accomplished by the use of statins. However, some patients do not respond sufficiently to statin therapy and therefore still have a residual cardiovascular risk. This issue highlights the need for novel therapeutic strategies. As macrophages are implicated in all stages of atherosclerotic lesion development, they represent an important alternative drug target. A variety of anti-inflammatory strategies have recently emerged to treat or prevent atherosclerosis. Here, we review the canonical mechanisms of macrophage death and their impact on atherogenesis and plaque stability. Macrophage death is a prominent feature of advanced plaques and is a major contributor to necrotic core formation and plaque destabilization. Mechanisms of macrophage death in atherosclerosis include apoptosis, passive or accidental necrosis as well as secondary necrosis, a type of death that typically occurs when apoptotic cells are insufficiently cleared by neighboring cells via a phagocytic process termed efferocytosis. In addition, less-well characterized types of regulated necrosis in macrophages such as necroptosis, pyroptosis, ferroptosis, and parthanatos may occur in advanced plaques and are also discussed. Autophagy in plaque macrophages is an important survival pathway that protects against cell death, yet massive stimulation of autophagy promotes another type of death, usually referred to as autosis. Multiple lines of evidence indicate that a better insight into the different mechanisms of macrophage death, and how they mutually interact, will provide novel pharmacological strategies to resolve atherosclerosis and stabilize vulnerable, rupture-prone plaques.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Isabelle Coornaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
17
|
The Crosstalk between Fat Homeostasis and Liver Regional Immunity in NAFLD. J Immunol Res 2019; 2019:3954890. [PMID: 30719457 PMCID: PMC6335683 DOI: 10.1155/2019/3954890] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/11/2018] [Accepted: 12/09/2018] [Indexed: 12/14/2022] Open
Abstract
The liver is well known as the center of glucose and lipid metabolism in the human body. It also functions as an immune organ. Previous studies have suggested that liver nonparenchymal cells are crucial in the progression of NAFLD. In recent years, NAFLD's threat to human health has been becoming a global issue. And by far, there is no effective treatment for NAFLD. Liver nonparenchymal cells are stimulated by lipid antigens, adipokines, or other factors, and secreted immune factors can alter the expression of key proteins such as SREBP-1c, ChREBP, and PPARγ to regulate lipid metabolism, thus affecting the pathological process of NAFLD. Interestingly, some ncRNAs (including miRNAs and lncRNAs) participate in the pathological process of NAFLD by changing body fat homeostasis. And even some ncRNAs could regulate the activity of HSCs, thereby affecting the progression of inflammation and fibrosis in the course of NAFLD. In conclusion, immunotherapy could be an effective way to treat NAFLD.
Collapse
|
18
|
Zong S, Li J, Yang L, Huang Q, Ye Z, Hou G, Ye M. Synergistic antitumor effect of polysaccharide from Lachnum sp. in combination with cyclophosphamide in hepatocellular carcinoma. Carbohydr Polym 2018; 196:33-46. [PMID: 29891303 DOI: 10.1016/j.carbpol.2018.05.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/28/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023]
Abstract
Combination therapy with chemotherapeutics is attracting increasing attention as an important treatment option for hepatocellular carcinoma (HCC) due to its complex pathological characteristics. In this study, as a new therapy strategy, combination treatment of LEP-2a (a non-toxic polysaccharide from Lachnum sp.) with cyclophosphamide (CTX) was investigated. Results showed that combination treatment with LEP-2a and CTX processed a significantly synergistic anti-tumor effect in H22 tumor-bearing mice through Fas/FasL mediated caspase-dependent death pathway and mitochondria apoptosis pathway. Moreover, our study indicated that LEP-2a played a crucial role in enhancement of immune response, inhibition of tumor angiogenesis and down-regulation of survival associated proteins. Notably, side effects induced by CTX were relieved after LEP-2a treatment. These results support the conception that LEP-2a has the potential as an ideal adjuvant agent for a more effective combination therapy with CTX against HCC.
Collapse
Affiliation(s)
- Shuai Zong
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Jinglei Li
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Liu Yang
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Qianli Huang
- School of Biological and Medical Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Ziyang Ye
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Guohua Hou
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Ming Ye
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
19
|
Peng Y, Jin J, Fan L, Xu H, He P, Li J, Chen T, Ruan W, Chen G. Rolipram Attenuates Early Brain Injury Following Experimental Subarachnoid Hemorrhage in Rats: Possibly via Regulating the SIRT1/NF-κB Pathway. Neurochem Res 2018; 43:785-795. [PMID: 29397536 DOI: 10.1007/s11064-018-2480-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/26/2022]
Abstract
Early brain injury (EBI) is the primary cause of poor outcome in subarachnoid hemorrhage (SAH) patients. Rolipram, a specific phosphodiesterase-4 inhibitor which is traditionally used as an anti-depressant drug, has been recently proven to exert neuroprotective effects in several central nervous system insults. However, the role of rolipram in SAH remains uncertain. The current study was aimed to investigate the role of rolipram in EBI after SAH and explore the potential mechanism. Adult male Sprague-Dawley rats were subjected to an endovascular perforation process to produce an SAH model. Rolipram was injected intraperitoneally at 2 h after SAH with a dose of 10 mg/kg. We found that rolipram significantly ameliorated brain edema and alleviated neurological dysfunction after SAH. Rolipram treatment remarkably promoted the expression of Sirtuin 1 (SIRT1) while inhibited NF-κB activation. Moreover, rolipram significantly inhibited the activation of microglia as well as down-regulated the expression of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6. In addition, rolipram increased the expression of protective cytokine IL-10. Furthermore, rolipram significantly alleviated neuronal death after SAH. In conclusion, these data suggested that rolipram exerts neuroprotective effects against EBI after SAH via suppressing neuroinflammation and reducing neuronal loss. The neuroprotective effects of rolipram were associated with regulating the SIRT1/NF-κB pathway. Rolipram could be a novel and promising therapeutic agent for SAH treatment.
Collapse
Affiliation(s)
- Yucong Peng
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianxiang Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Linfeng Fan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Hangzhe Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Pingyou He
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Jianru Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Ting Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Wu Ruan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China
| | - Gao Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Jiefang Road 88th, Hangzhou, 310016, China.
| |
Collapse
|