1
|
Guo S, Zhang Y, Lian J, Su C, Wang H. The role of hydrogen sulfide in the regulation of necroptosis across various pathological processes. Mol Cell Biochem 2025; 480:1999-2013. [PMID: 39138751 DOI: 10.1007/s11010-024-05090-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Necroptosis is a programmed cell death form executed by receptor-interacting protein kinase (RIPK) 1, RIPK3 and mixed lineage kinase domain-like protein (MLKL), which assemble into an oligomer called necrosome. Accumulating evidence reveals that necroptosis participates in many types of pathological processes. Hence, clarifying the mechanism of necroptosis in pathological processes is particularly important for the prevention and treatment of various diseases. For over 300 years, hydrogen sulfide (H2S) has been widely known in the scientific community as a toxic and foul-smelling gas. However, after discovering the important physiological and pathological functions of H2S, human understanding of this small molecule changed, believing that H2S is the third gas signaling molecule after carbon monoxide (CO) and nitric oxide (NO). H2S plays an important role in various diseases, but the related mechanisms are not yet fully understood. In recent years, more and more studies have shown that H2S regulation of necroptosis is involved in various pathological processes. Herein, we focus on the recent progress on the role of H2S regulation of necroptosis in different pathological processes and profoundly analyze the related mechanisms.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Yanting Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Jingwen Lian
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Chunqi Su
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Honggang Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
2
|
Han L, Zhai W. Mechanisms and preventive measures of ALDH2 in ischemia‑reperfusion injury: Ferroptosis as a novel target (Review). Mol Med Rep 2025; 31:105. [PMID: 40017132 DOI: 10.3892/mmr.2025.13470] [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: 10/17/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
Ischemia‑reperfusion injury (IRI) refers to tissue or organ damage that occurs following a period of inadequate blood supply (ischemia) followed by restoration of blood flow (reperfusion) within a short time frame. This phenomenon is prevalent in clinical conditions such as cardiovascular and cerebrovascular disease, organ transplantation and stroke. Despite its frequency, effective therapeutic strategies to mitigate IRI remain elusive in clinical practice, underscoring the need for a deeper understanding of its molecular mechanisms. Aldehyde dehydrogenase 2 (ALDH2), a key enzyme in alcohol metabolism, serves a role in alleviating oxidative stress and cell damage during IRI by modulating oxidative stress, decreasing apoptosis and inhibiting inflammatory responses. ALDH2 exerts protective effects by detoxifying reactive aldehydes, thereby preventing lipid peroxidation and maintaining cellular homeostasis. Furthermore, ferroptosis, a regulated form of cell death driven by iron accumulation and subsequent lipid peroxidation, is a key process in IRI. However, the precise role of ALDH2 in modulating ferroptosis during IRI remains incompletely understood. Although there is an interaction between ALDH2 activity and ferroptosis, the underlying mechanisms have yet to be clarified. The present review examines the role of ALDH2 and ferroptosis in IRI and the potential regulatory influence of ALDH2 on ferroptosis mechanisms, as well as potential targeting of ALDH2 and ferroptosis for IRI treatment and prevention. By elucidating the complex interplay between ALDH2 and ferroptosis, the present review aims to provide new insights for the development of innovative therapeutic strategies to mitigate ischemic tissue damage and improve clinical outcomes.
Collapse
Affiliation(s)
- Liang Han
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| | - Wen Zhai
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
3
|
Jiang P, Chipurupalli S, Yoo BH, Liu X, Rosen KV. Inactivation of necroptosis-promoting protein MLKL creates a therapeutic vulnerability in colorectal cancer cells. Cell Death Dis 2025; 16:118. [PMID: 39979285 PMCID: PMC11842741 DOI: 10.1038/s41419-025-07436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Mortality from colorectal cancer (CRC) is significant, and novel CRC therapies are needed. A pseudokinase MLKL typically executes necroptotic cell death, and MLKL inactivation protects cells from such death. However, we found unexpectedly that MLKL gene knockout enhanced CRC cell death caused by a protein synthesis inhibitor homoharringtonine used for chronic myeloid leukemia treatment. In an effort to explain this finding, we observed that MLKL gene knockout reduces the basal CRC cell autophagy and renders such autophagy critically dependent on the presence of VPS37A, a component of the ESCRT-I complex. We further found that the reason why homoharringtonine enhances CRC cell death caused by MLKL gene knockout is that homoharringtonine activates p38 MAP kinase and thereby prevents VPS37A from supporting autophagy in MLKL-deficient cells. We observed that the resulting inhibition of the basal autophagy in CRC cells triggers their parthanatos, a cell death type driven by poly(ADP-ribose) polymerase hyperactivation. Finally, we discovered that a pharmacological MLKL inhibitor necrosulfonamide strongly cooperates with homoharringtonine in suppressing CRC cell tumorigenicity in mice. Thus, while MLKL promotes cell death during necroptosis, MLKL supports the basal autophagy in CRC cells and thereby protects them from death. MLKL inactivation reduces such autophagy and renders the cells sensitive to autophagy inhibitors, such as homoharringtonine. Hence, MLKL inhibition creates a therapeutic vulnerability that could be utilized for CRC treatment.
Collapse
Affiliation(s)
- Peijia Jiang
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Sandhya Chipurupalli
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Byong Hoon Yoo
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Xiaoyang Liu
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Kirill V Rosen
- Departments of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
4
|
Gonçalves J, Amaral JD, Capela R, Perry MDJ, Braga C, Gaspar MM, Piedade FM, Bijlsma L, Roig A, Pinto SN, Moreira R, Florindo P, Rodrigues CMP. Necroptosis induced by ruthenium (II) complexes as mitochondrial disruptors. Cell Death Discov 2024; 10:261. [PMID: 38806468 PMCID: PMC11133381 DOI: 10.1038/s41420-024-02033-z] [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: 01/20/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
Inducing necroptosis in cancer cells has emerged as an effective strategy to overcome drug resistance. However, while organic small molecules have been extensively studied for this purpose, metal-based compounds have received relatively little attention as triggers of necroptosis. The development of ruthenium (II) hybrid compounds, particularly those containing triazene (Ru-TRZ), highlights a novel avenue for modulating necroptotic cell death. Here we show that incorporating a methyltriazene moiety, a known alkylating warhead, confers superior mitochondrial-targeting properties and enhances cell death compared to amide-containing counterparts. Ru-hybrid TRZ2 exhibits also antitumor efficacy against in vivo drug-resistant cancer cells. Mechanistically, we demonstrate that Ru-TRZ hybrids induce apoptosis. In addition, by activating downstream RIPK3-driven cell death, TRZ2 proficiently restrains normal mitochondrial function and activity, leading to cancer cell necroptosis. Finally, TRZ2 synergizes anti-proliferative activity and cell death effects induced by conventional drugs. In conclusion, Ru-TRZ2 stands as a promising ruthenium-based chemotherapeutic agent inducing necroptosis in drug resistant cancer cells.
Collapse
Affiliation(s)
- Joana Gonçalves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana D Amaral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Rita Capela
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria de Jesus Perry
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cláudia Braga
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Instituto de Biofísica e Engenharia Biomédica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal
| | - Fátima M Piedade
- Departamento de Química e Bioquímica, Faculty of Sciences, Universidade de Lisboa, Lisbon, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Lubertus Bijlsma
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, Castelló, Spain
| | - Antoni Roig
- Environmental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, Castelló, Spain
| | - Sandra N Pinto
- iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Rui Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
5
|
Liu S, Liu C, Wang Y, Chen J, He Y, Hu K, Li T, Yang J, Peng J, Hao L. The role of programmed cell death in osteosarcoma: From pathogenesis to therapy. Cancer Med 2024; 13:e7303. [PMID: 38800967 PMCID: PMC11129166 DOI: 10.1002/cam4.7303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/01/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Osteosarcoma (OS) is a prevalent bone solid malignancy that primarily affects adolescents, particularly boys aged 14-19. This aggressive form of cancer often leads to deadly lung cancer due to its high migration ability. Experimental evidence suggests that programmed cell death (PCD) plays a crucial role in the development of osteosarcoma. Various forms of PCD, including apoptosis, ferroptosis, autophagy, necroptosis, and pyroptosis, contribute significantly to the progression of osteosarcoma. Additionally, different signaling pathways such as STAT3/c-Myc signal pathway, JNK signl pathway, PI3k/AKT/mTOR signal pathway, WNT/β-catenin signal pathway, and RhoA signal pathway can influence the development of osteosarcoma by regulating PCD in osteosarcoma cell. Therefore, targeting PCD and the associated signaling pathways could offer a promising therapeutic approach for treating osteosarcoma.
Collapse
Affiliation(s)
- Suqing Liu
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Chengtao Liu
- Shandong Wendeng Osteopathic HospitalWeihaiChina
| | - Yian Wang
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Jiewen Chen
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Yujin He
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Kaibo Hu
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Ting Li
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Junmei Yang
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jie Peng
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Sports Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Liang Hao
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
6
|
Liu S, Huang B, Cao J, Wang Y, Xiao H, Zhu Y, Zhang H. ROS fine-tunes the function and fate of immune cells. Int Immunopharmacol 2023; 119:110069. [PMID: 37150014 DOI: 10.1016/j.intimp.2023.110069] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023]
Abstract
The redox state is essential to the process of cell life, which determines cell fate. As an important signaling molecule of the redox state, reactive oxygen species (ROS) are crucial for the homeostasis of immune cells and participate in the pathological processes of different diseases. We discuss the underlying mechanisms and possible signaling pathways of ROS to fine-tune the proliferation, differentiation, polarization and function of immune cells, including T cells, B cells, neutrophils, macrophages, myeloid-derived inhibitory cells (MDSCs) and dendritic cells (DCs). We further emphasize how excessive ROS lead to programmed immune cell death such as apoptosis, ferroptosis, pyroptosis, NETosis and necroptosis, providing valuable insights for future therapeutic strategies in human diseases.
Collapse
Affiliation(s)
- Shiyu Liu
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Benqi Huang
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Jingdong Cao
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Yifei Wang
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Hao Xiao
- Department of Clinical Medicine, Xiangya School of Medicine, Central South University, 410008 Changsha, China
| | - Yaxi Zhu
- Sepsis Translational Medicine Key Lab of Hunan Province, Department of Pathophysiology, School of Basic Medical Sciences, Central South University, 410008 Changsha, China.
| | - Huali Zhang
- Sepsis Translational Medicine Key Lab of Hunan Province, Department of Pathophysiology, School of Basic Medical Sciences, Central South University, 410008 Changsha, China.
| |
Collapse
|
7
|
Zhang W, Sun X, Shi X, Qi X, Shang S, Lin H. Subacute Cadmium Exposure Induces Necroptosis in Swine Lung via Influencing Th1/Th2 Balance. Biol Trace Elem Res 2023; 201:220-228. [PMID: 35118606 DOI: 10.1007/s12011-022-03133-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/23/2022] [Indexed: 01/11/2023]
Abstract
Cadmium (Cd) is a type of toxic substance, which widely exists in nature. However, the effect of Cd exposure on the toxicity of swine lungs and its underlying mechanism involved have not yet been reported. In our study, we divided swine into two groups, including a control group (C group) and Cd-exposed group. Swine in the C group were fed a basic diet, whereas swine in the Cd group were fed a 20 mg Cd/kg diet. Immunofluorescence, qRT-PCR, western blot analysis, and H&E staining were performed to detect necroptosis-related indicators. Our results found that after Cd exposure, Th1/Th2 imbalance occurred, miR-181-5p was down-regulated, TNF-α expression was increased, and the NF-κB/NLRP3 and JAK/STAT pathways and RIPK1/RIPK3/MLKL axis were activated. Furthermore, histopathological examination showed necrosis in swine lung after Cd exposure. Together, the above-mentioned results indicate that subacute Cd exposure is closely linked with necroptosis in swine lung. Our study provided evidence that Cd may act through miR-181-5p/TNF-α to induce necroptosis in swine lung. The findings of this study supplement the toxicological study of Cd and provide a reference for comparative medicine.
Collapse
Affiliation(s)
- Wenyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xinyue Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Xue Qi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Shaoqian Shang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, People's Republic of China.
| |
Collapse
|
8
|
Li X, Zhang Y, Wang J, Li Y, Wang Y, Shi F, Hong L, Li L, Diao H. zVAD alleviates experimental autoimmune hepatitis in mice by increasing the sensitivity of macrophage to TNFR1-dependent necroptosis. J Autoimmun 2022; 133:102904. [PMID: 36108506 DOI: 10.1016/j.jaut.2022.102904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Autoimmune hepatitis (AIH) is characterized by hepatocyte destruction, leading to lymphocyte and macrophage accumulation in the liver. Macrophages are key drivers of AIH. A membrane-permeable pan-caspase inhibitor, Z-Val-Ala-DL-Asp-fluoromethylketone (zVAD), induces macrophage necroptosis in response to certain stimuli. However, the function of zVAD in the pathogenesis of autoimmune hepatitis remains elusive. In this study, we aimed to evaluate the effect and explore the underlying mechanisms of zVAD against AIH. METHODS Murine acute autoimmune liver injury was established by concanavalin A (ConA) injection. Bone marrow-derived macrophages (BMDMs) were used in adoptive cell transfer experiments. The mechanism of action of zVAD was examined using macrophage cell lines and BMDMs. Phosphorylation of mixed lineage kinase domain-like proteins was used as a marker of necroptosis. RESULTS Treatment with zVAD increased necroptosis, reduced inflammatory cytokine production, and alleviated liver injury in a ConA-induced liver injury mouse model. Regardless of zVAD treatment, macrophage deletion resulted in reduced neutrophil accumulation and relieved ConA-induced liver injury. In vitro studies have shown that zVAD pretreatment promotes lipopolysaccharide-induced macrophage necroptosis and leads to reduced pro-inflammatory cytokine and chemokine secretion. Transferring zVAD-pretreated BMDMs in mice notably reduced ConA-associated liver inflammation and injury, resulting in lower mortality than that observed after transferring normal BMDMs. Mechanistically, zVAD treatment increased the expression of tumour necrosis factor receptor (TNFR)-1 and interleukin (IL)-10 in macrophages. TNFR1 expression decreased upon transfection with IL-10-specific small interfering RNAs and blocking of TNFR1 decreased macrophage necroptosis. CONCLUSIONS We found that zVAD alleviated ConA-induced liver injury by increasing the sensitivity of macrophages to necroptosis via IL-10-induced TNFR1 expression. This study provides new insights into the treatment of autoimmune hepatitis via zVAD-induced macrophage necroptosis.
Collapse
Affiliation(s)
- Xuehui Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongting Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinping Wang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yuyu Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuchong Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Hong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
9
|
Gupta P, Singh A, Verma AK, Kant S, Pandey AK, Khare P, Prakash V. The Anti-Tumor and Immunomodulatory Effects of PLGA-Based Docetaxel Nanoparticles in Lung Cancer: The Potential Involvement of Necroptotic Cell Death through Reactive Oxygen Species and Calcium Build-Up. Vaccines (Basel) 2022; 10:1801. [PMID: 36366309 PMCID: PMC9694303 DOI: 10.3390/vaccines10111801] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/05/2022] [Accepted: 10/19/2022] [Indexed: 04/23/2025] Open
Abstract
Taxanes, microtubule stabilizing agents, are extensively used in the treatment of non-small cell lung cancer (NSCLC). However, their clinical effectiveness remains restricted owing to significant adverse effects and drug resistance. Nanotechnology may guide chemotherapeutic drugs directly and selectively to malignant cells, improving their therapeutic efficiency. In the present study, we synthesized polylactic-co-glycolic acid (PLGA) based nanoparticles encapsulating docetaxel and evaluated their efficacy in non-small cell lung carcinoma (A549) cells and primary immune cells derived from humans. Docetaxel-PLGA nanoparticles (PLGA-Dtx) were synthesized and characterized using distinct methods. Moreover, the cytotoxicity of free docetaxel (Dtx) and Dtx-conjugated nanoparticles (PLGA-Dtx) was studied in A549 cells and peripheral blood mononuclear cells derived from humans. Furthermore, annexin V-FITC/PI staining was used to assess the mode of cell death. Additionally, human peripheral blood mononuclear cells (PBMCs) were used for assessing the associated immune response and cytokine profile following PLGA-Dtx treatment. Spherical PLGA-Dtx nanoparticles with a 150 ± 10 nm diameter and 70% encapsulation efficiency (EE) were synthesized. The MTT assay showed that the IC50 of PLGA-Dtx nanoparticles was significantly lower than free docetaxel in A549 cells. Cytotoxicity data also revealed the selective nature of PLGA-Dtx with no significant effects in normal human bronchial epithelial cells (BEAS-2B) and PBMCs derived from healthy donors. Interestingly, PLGA-Dtx exerted an improved effect and tempted both apoptosis and necroptosis, as evidenced by annexin V and propidium iodide-positive cells. Further, PLGA-Dtx-exposed A549 cells showed increased Cas-3, Cas-9, RIP-1, and RIP-3, indicating apoptosis and necroptosis. An increased pro-inflammatory response manifested from the enhancement of IFN-γ and TNF-α in PLGA-Dtx-exposed PBMCs, posed by the occurrence of necroptosis and the immune stimulatory effect of PLGA-Dtx. In conclusion, PLGA-Dtx has a selective anticancer potential and better immunostimulatory potential. Therefore, PLGA-Dtx may be useful for the treatment of non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Arpita Singh
- Department of Pharmacology, Dr. Ram Manohar Lohia, Institute of Medical Sciences, Lucknow 226010, Uttar Pradesh, India
| | - Ajay Kumar Verma
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Surya Kant
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Anuj Kumar Pandey
- Department of Respiratory Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Puneet Khare
- Flow Cytometry Facility, Central Instrumentation Facility, CSIR-Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India
| | - Ved Prakash
- Department of Pulmonary & Critical Care Medicine, King George’s Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
10
|
Abdelbary M, Mohamed R, Gillis EE, Diaz-Sanders K, Baban B, Brands MW, Sullivan JC. Sex differences in apoptosis do not contribute to sex differences in blood pressure or renal T cells in spontaneously hypertensive rats. Front Physiol 2022; 13:1006951. [PMID: 36304583 PMCID: PMC9592703 DOI: 10.3389/fphys.2022.1006951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Apoptosis is a physiological and anti-inflammatory form of cell death that is indispensable for normal physiology and homeostasis. Several studies have reported aberrant activation of apoptosis in various tissues at the onset of hypertension. However, the functional significance of apoptosis during essential hypertension remains largely undefined. The current study was designed to test the hypothesis that apoptosis contributes to sex differences in blood pressure and the T cell profile in spontaneously hypertensive rats (SHR). Apoptosis was measured in kidney, aorta and spleen of 13-week-old adult hypertensive male and female SHR. Female SHR had greater renal and aortic apoptosis compared to age-matched males; apoptosis in the spleen was comparable between the sexes. Based on well-established sex differences in hypertension, we tested the hypothesis that greater apoptosis in female SHR contributes to the lower BP and pro-inflammatory profile compared to males. Male and female SHR were randomized to receive vehicle or ZVAD-FMK, a cell permeable pan-caspase inhibitor, in established hypertension from 13 to 15 weeks of age or at the onset of hypertension from 6 to 12 weeks or age. Treatment with ZVAD-FMK lowered renal apoptosis in both studies, yet neither BP nor renal T cells were altered in either male or female SHR. These results suggest that apoptosis does not contribute to the control or maintenance of BP in male or female SHR or sex differences in renal T cells.
Collapse
Affiliation(s)
- Mahmoud Abdelbary
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Riyaz Mohamed
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Ellen E. Gillis
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Karl Diaz-Sanders
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Babak Baban
- Department of Oral Biology, Augusta University, Augusta, GA, United States
| | - Michael W. Brands
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Jennifer C. Sullivan
- Department of Physiology, Augusta University, Augusta, GA, United States,*Correspondence: Jennifer C. Sullivan,
| |
Collapse
|
11
|
The regulation of necroptosis and perspectives for the development of new drugs preventing ischemic/reperfusion of cardiac injury. Apoptosis 2022; 27:697-719. [DOI: 10.1007/s10495-022-01760-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/11/2022]
|
12
|
Panahipour L, Cervantes LCC, Oladzad Abbasabadi A, Sordi MB, Kargarpour Z, Gruber R. Blocking of Caspases Exerts Anti-Inflammatory Effects on Periodontal Cells. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071045. [PMID: 35888133 PMCID: PMC9316350 DOI: 10.3390/life12071045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022]
Abstract
Periodontitis is an inflammatory process that is associated with caspase activity. Caspases could thus become molecular targets for the modulation of the inflammatory response to harmful factors, such as lipopolysaccharides (LPS) and TNFα. Here, the impact of the pan-caspase inhibitor Z-VAD-FMK (carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoro-methyl ketone) on the modulation of the LPS-induced inflammatory response of murine RAW 264.7 cells and primary macrophages was examined. Moreover, the inflammatory responses of human gingival fibroblasts, HSC2 oral squamous carcinoma cells and murine ST2 mesenchymal fibroblasts when exposed to TNFα were studied. Data showed that Z-VAD-FMK significantly lowered the inflammatory response of RAW 264.7 cells and primary macrophages, as indicated by the expression of IL1 and IL6. In murine ST2 mesenchymal fibroblasts, the TNFα-induced expression of CCL2 and CCL5 was significantly reduced. In human gingival fibroblasts and HSC2 cells, Z-VAD-FMK considerably reduced the TNFα-induced expression of CXCL8 and CXCL10. These findings suggest that pharmacological blocking of caspases in an inflammatory environment lowers the expression of cytokines and chemokines in periodontal cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Lara Cristina Cunha Cervantes
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Sao Paulo 16015-050, Brazil
| | - Azarakhsh Oladzad Abbasabadi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Mariane Beatriz Sordi
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Centre for Research on Dental Implants (CEPID), Department of Dentistry, Federal University of Santa Catarina (UFSC), Florianopolis 88040-900, Brazil
| | - Zahra Kargarpour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (L.C.C.C.); (A.O.A.); (M.B.S.); (Z.K.)
- Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200 Vienna, Austria
- Correspondence:
| |
Collapse
|
13
|
Verduijn J, Van der Meeren L, Krysko DV, Skirtach AG. Deep learning with digital holographic microscopy discriminates apoptosis and necroptosis. Cell Death Dis 2021; 7:229. [PMID: 34475384 PMCID: PMC8413278 DOI: 10.1038/s41420-021-00616-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
Regulated cell death modalities such as apoptosis and necroptosis play an important role in regulating different cellular processes. Currently, regulated cell death is identified using the golden standard techniques such as fluorescence microscopy and flow cytometry. However, they require fluorescent labels, which are potentially phototoxic. Therefore, there is a need for the development of new label-free methods. In this work, we apply Digital Holographic Microscopy (DHM) coupled with a deep learning algorithm to distinguish between alive, apoptotic and necroptotic cells in murine cancer cells. This method is solely based on label-free quantitative phase images, where the phase delay of light by cells is quantified and is used to calculate their topography. We show that a combination of label-free DHM in a high-throughput set-up (~10,000 cells per condition) can discriminate between apoptosis, necroptosis and alive cells in the L929sAhFas cell line with a precision of over 85%. To the best of our knowledge, this is the first time deep learning in the form of convolutional neural networks is applied to distinguish-with a high accuracy-apoptosis and necroptosis and alive cancer cells from each other in a label-free manner. It is expected that the approach described here will have a profound impact on research in regulated cell death, biomedicine and the field of (cancer) cell biology in general.
Collapse
Affiliation(s)
- Joost Verduijn
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Louis Van der Meeren
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Dmitri V. Krysko
- grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium ,grid.5342.00000 0001 2069 7798Cell Death Investigation and Therapy (CDIT) Laboratory, Anatomy an Embryology Unit, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium ,grid.448878.f0000 0001 2288 8774Department of Pathophysiology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russian Federation
| | - André G. Skirtach
- grid.5342.00000 0001 2069 7798Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium ,grid.510942.bCancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
14
|
Wang F, Tang L, Liang B, Jin C, Gao L, Li Y, Li Z, Shao J, Zhang Z, Tan S, Zhang F, Zheng S. Yi-Qi-Jian-Pi Formula Suppresses RIPK1/RIPK3-Complex-Dependent Necroptosis of Hepatocytes Through ROS Signaling and Attenuates Liver Injury in Vivo and in Vitro. Front Pharmacol 2021; 12:658811. [PMID: 33967802 PMCID: PMC8102982 DOI: 10.3389/fphar.2021.658811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Acute-on-chronic liver failure (ACLF) is described as a characteristic of acute jaundice and coagulation dysfunction. Effective treatments for ACLF are unavailable and hence are urgently required. We aimed to define the effect of Yi-Qi-Jian-Pi Formula (YQJPF) on liver injury and further examine the molecular mechanisms. In this study, we established CCl4-, LPS-, and d-galactosamine (D-Gal)-induced ACLF rat models in vivo and LPS- and D-Gal-induced hepatocyte injury models in vitro. We found that YQJPF significantly ameliorates liver injury in vivo and in vitro that is associated with the regulation of hepatocyte necroptosis. Specifically, YQJPF decreased expression of receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and pseudokinase mixed lineage kinase domain-like (MLKL) to inhibit the migration of RIPK1 and RIPK3 into necrosome. YQJPF also reduces the expression of inflammatory cytokines IL-6, IL-8, IL-1β, and TNF-α, which were regulated by RIPK3 mediates cell death. RIPK1 depletion was found to enhance the protective effect of YQJPF. Furthermore, we showed that YQJPF significantly downregulates the mitochondrial reactive oxygen species (ROS) production and mitochondrial depolarization, with ROS scavenger, 4-hydroxy-TEMPO treatment recovering impaired RIPK1-mediated necroptosis and reducing the expression of IL-6, IL-8, IL-1β, and TNF-α. In summary, our study revealed the molecular mechanism of protective effect of YQJPF on hepatocyte necroptosis, targeting RIPK1/RIPK3-complex-dependent necroptosis via ROS signaling. Overall, our results provided a novel perspective to indicate the positive role of YQJPF in ACLF.
Collapse
Affiliation(s)
- Feixia Wang
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Tang
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Baoyu Liang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chun Jin
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liyuan Gao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhanghao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shanzhong Tan
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
15
|
Peng L, Wang Y, Yang B, Qin Q, Song E, Song Y. Polychlorinated biphenyl quinone regulates MLKL phosphorylation that stimulates exosome biogenesis and secretion via a short negative feedback loop. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 274:115606. [PMID: 33190980 DOI: 10.1016/j.envpol.2020.115606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/06/2020] [Accepted: 09/01/2020] [Indexed: 06/11/2023]
Abstract
Polychlorinated biphenyls (PCBs) are one of the most refractory organic environmental pollutants that ubiquitous existence in nature. Due to the polymorphism of their metabolic pathway and corresponding downstream metabolites, PCBs' toxicities are complicated and need extended investigation. In the present study, we discovered a novel regulatory mechanism of PCB quinone metabolite-driven programmed cell death (PCD), namely, necroptosis. We first confirmed that PCB quinone induces cancerous HeLa and MDA-MB-231 cells necroptosis via the phosphorylation of mixed lineage kinase domain-like MLKL (p-MLKL). Then, we found that PCB quinone-stimulated p-MLKL enhances exosome biogenesis and secretion. Exosome interacts with p-MLKL and releases p-MLKL to the outside of the cell, and ultimately alleviating PCB quinone-induced necroptosis. The inhibition of exosome secretion by GW4869 significantly elevated necroptotic level, indicating the establishment of a short negative feedback loop of MLKL-exosome secretion upon PCB quinone challenge. Since exosome-mediated signaling showed great implications in various human diseases, this work may provide a new mechanism for PCBs-associated toxicity.
Collapse
Affiliation(s)
- Lu Peng
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yawen Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Bingwei Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Qi Qin
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
16
|
Koike A, Tsujinaka K, Fujimori K. Statins attenuate antiviral IFN-β and ISG expression via inhibition of IRF3 and JAK/STAT signaling in poly(I:C)-treated hyperlipidemic mice and macrophages. FEBS J 2021; 288:4249-4266. [PMID: 33452755 DOI: 10.1111/febs.15712] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/07/2020] [Accepted: 01/13/2021] [Indexed: 12/17/2022]
Abstract
Viral infection is a significant burden to health care worldwide. Statins, 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibitors, are widely used as cholesterol-lowering drugs. Recently, long-term statin therapy was shown to reduce the antiviral immune response; however, the underlying molecular mechanisms are unclear. Here, we found that simvastatin decreased polyinosinic-polycytidylic acid [poly(I:C)]-induced expression of antiviral interferon (IFN)-β and IFN-stimulated genes (ISGs) in the bronchoalveolar lavage fluid (BALF) and lungs of mice with high-fat diet-induced hyperlipidemia. Macrophages were the dominant cell type in the BALF of poly(I:C)-treated mice. We examined the effects of simvastatin in primary lung macrophages and found that simvastatin suppressed poly(I:C)-induced expression of IFN-β and ISGs. We examined the molecular mechanisms of statin-mediated inhibition of antiviral gene expression using murine macrophage-like cell line, J774.1/JA-4. Simvastatin and pitavastatin decreased poly(I:C)-induced expression of IFN-β and ISGs. Moreover, they repressed poly(I:C)-induced phosphorylation of IFN regulatory factor (IRF) 3 and signal transducers and activators of transcription (STAT) 1, which is involved in Janus kinase (JAK)/STAT signaling. Mevalonate and geranylgeranyl pyrophosphate (GGPP), but not cholesterol, counteracted the negative effect of statins on IFN-β and ISG expression and phosphorylation of IRF3 and STAT1. The geranylgeranyltransferase inhibitor suppressed poly(I:C)-induced expression of IFN-β and ISGs and phosphorylation of IRF3 and STAT1. These results suggest that statins suppressed the expression of IFN-β and ISGs in poly(I:C)-treated hyperlipidemic mice and murine macrophages and that these effects occurred through the inhibition of IRF3 and JAK/STAT signaling in macrophages. Furthermore, GGPP recovered the statin-suppressed IRF3 and JAK/STAT signaling in poly(I:C)-treated macrophages.
Collapse
Affiliation(s)
- Atsushi Koike
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan
| | - Kaito Tsujinaka
- Osaka University of Pharmaceutical Sciences, Takatsuki, Japan
| | - Ko Fujimori
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences, Takatsuki, Japan
| |
Collapse
|
17
|
Lin J, Li X, Lin Y, Huang Z, Wu W. Exogenous sodium hydrosulfide protects against high glucose‑induced injury and inflammation in human umbilical vein endothelial cells by inhibiting necroptosis via the p38 MAPK signaling pathway. Mol Med Rep 2021; 23:67. [PMID: 33215220 PMCID: PMC7716414 DOI: 10.3892/mmr.2020.11706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/05/2020] [Indexed: 12/30/2022] Open
Abstract
In recent years hydrogen sulfide (H2S) has demonstrated vasculoprotective effects against cell death, which suggests its promising therapeutic potential for numerous types of disease. Additionally, a protective effect of exogenous H2S in HG‑induced injuries in HUVECs was demonstrated, suggesting a potential protective effect for diabetic vascular complications. The present study aimed to investigate the mechanism accounting for the cytoprotective role of exogenous H2S against high glucose [HG (40 mM glucose)]‑induced injury and inflammation in human umbilical vein endothelial cells (HUVECs). HUVECs were exposed to HG for 24 h to establish an in vitro model of HG‑induced cytotoxicity. The cells were pretreated with sodium hydrosulfide (NaHS), a donor of H2S, or inhibitors of necroptosis and p38 MAPK prior to the exposure to HG. Cell viability, intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP), IL‑1β, IL‑6, IL‑8, TNF‑α, phosphorylated‑(p)38 and receptor‑interacting protein 3 (RIP3) expression levels were detected using the indicated methods, including Cell Counting Kit 8, fluorescence detection, western blotting, immunofluorescence assay and ELISAs. The results demonstrated that necroptosis and the p38 MAPK signaling pathway mediated HG‑induced injury and inflammation. Notably, NaHS was discovered to significantly ameliorate p38 MAPK/necroptosis‑mediated injury and inflammation in response to HG, as evidenced by an increase in cell viability, a decrease in ROS generation and loss of MMP, as well as the reduction in the secretion of proinflammatory cytokines. In addition, the upregulated expression of RIP3 induced by HG was repressed by treatment with SB203580, while the HG‑induced upregulation of p‑p38 expression levels were significantly downregulated following the treatment of Nec‑1 and RIP3‑siRNA. In conclusion, the findings of the present study indicated that NaHS may protect HUVECs against HG‑induced injury and inflammation by inhibiting necroptosis via the p38 MAPK signaling pathway, which may represent a promising drug for the therapy of diabetic vascular complications.
Collapse
Affiliation(s)
- Jiaqiong Lin
- Department of Endocrinology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoyong Li
- Department of Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Yan Lin
- Department of Nephrology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510150, P.R. China
| | - Zena Huang
- Department of General Medicine, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| | - Wen Wu
- Department of Endocrinology, Guangdong Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
18
|
Koike A. [Molecular Mechanism Underlying Inflammatory Cell Death via Necroptosis in M1 Macrophages]. YAKUGAKU ZASSHI 2020; 140:1427-1432. [PMID: 33268684 DOI: 10.1248/yakushi.20-00161] [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: 11/22/2022]
Abstract
M1 macrophages, also known as inflammatory macrophages, play an important role in the innate and adaptative immune responses against pathogens. However, the overactivation of these macrophages leads to the development and progression of various inflammatory diseases. Thus, the regulation of these macrophages is necessary to prevent such diseases. Necroptosis, a programmed form of necrosis, induces several damage-associated molecular patterns, such as high-mobility group box 1, adenosine triphosphate, and mitochondrial DNA, which activate various immune cells, thus leading to inflammation. Recent studies have shown that necroptosis in M1 macrophages is associated with inflammation in many pathological conditions. However, the molecular mechanisms underlying necroptosis in M1 macrophages are not completely understood. Thus, we examined the effects of a broad-spectrum caspase inhibitor, zVAD-fmk, on cell death in lipopolysaccharide (LPS)-induced M1 macrophages. Necrostatin-1, an inhibitor of necroptosis, partially inhibited zVAD-fmk-induced cell death and phosphorylation of mixed lineage kinase domain-like protein (MLKL) in M1 macrophages. Moreover, the inhibition of generation of reactive oxygen species (ROS) and activation of p38 mitogen-activated protein kinase (MAPK) reduced zVAD-fmk-induced necroptosis in M1 macrophages. Furthermore, the inhibition of ROS generation suppressed the activation of MLKL and p38 MAPK in zVAD-fmk-treated M1 macrophages. These results indicate that zVAD-fmk-induced cell death occurs via necroptosis through ROS-mediated activation of MLKL and p38 MAPK in M1 macrophages. Unraveling the molecular mechanisms of necroptosis in M1 macrophages might help understand their significance in inflammatory diseases.
Collapse
Affiliation(s)
- Atsushi Koike
- Department of Pathobiochemistry, Osaka University of Pharmaceutical Sciences
| |
Collapse
|
19
|
Wu Y, Dong G, Sheng C. Targeting necroptosis in anticancer therapy: mechanisms and modulators. Acta Pharm Sin B 2020; 10:1601-1618. [PMID: 33088682 PMCID: PMC7563021 DOI: 10.1016/j.apsb.2020.01.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/19/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023] Open
Abstract
Necroptosis, a genetically programmed form of necrotic cell death, serves as an important pathway in human diseases. As a critical cell-killing mechanism, necroptosis is associated with cancer progression, metastasis, and immunosurveillance. Targeting necroptosis pathway by small molecule modulators is emerging as an effective approach in cancer therapy, which has the advantage to bypass the apoptosis-resistance and maintain antitumor immunity. Therefore, a better understanding of the mechanism of necroptosis and necroptosis modulators is necessary to develop novel strategies for cancer therapy. This review will summarize recent progress of the mechanisms and detecting methods of necroptosis. In particular, the relationship between necroptosis and cancer therapy and medicinal chemistry of necroptosis modulators will be focused on.
Collapse
|
20
|
Kang P, Wang J, Fang D, Fang T, Yu Y, Zhang W, Shen L, Li Z, Wang H, Ye H, Gao Q. Activation of ALDH2 attenuates high glucose induced rat cardiomyocyte fibrosis and necroptosis. Free Radic Biol Med 2020; 146:198-210. [PMID: 31689484 DOI: 10.1016/j.freeradbiomed.2019.10.416] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/03/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Necroptosis is one of a regulated programmed death mode, fibrosis is closely related with cell death. It has been reported that inhibition of necroptosis can play the protective role in cardiac ischemia and reperfusion injury, stroke and other diseases, but the mechanisms of aldehyde dehydrogenases 2 (ALDH2) against high glucose induced neonatal rat ventricular primary cardiomyocytes fibrosis and necroptosis had not been elucidated clearly. This study was to observe the effect of ALDH2 on high glucose (HG) induced myocardial fibrosis and necroptosis in primary rat cardiomyocytes model. In contrast to normal glucose group, in HG group, with the decreases of ALDH2 activity, mRNA and protein levels, the cardiomyocytes viability was decreased, reactive oxygen species (ROS), the inflammation factors - tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1β (IL-1β) levels, collagen I (col I) and col III mRNA expressions and tissue inhibitors of matrix metalloproteinase 4 (TIMP4) protein expression were increased, while matrix metalloproteinase 14 (MMP14) protein level, the ratio of MMP14/TIMP4 were decreased, and the necroptosis key factors - the receptor interacting protein 1 (RIP1), RIP3 and mixed lineage kinase domain-like protein (MLKL) at mRNA and protein expressions were increased, the inflammasome core proteins - NLRP3 and ASC protein expressions were also increased, the apoptosis rate and necrosis rate were also increased. When the cardiomyocytes were treated with Alda-1 (the ALDH2 agonist) in HG intervention, the cell viability, ALDH2 activity, mRNA and protein levels, MMP14 protein level, the ratio of MMP14/TIMP4 were higher, ROS and TNF-α, IL-6, IL-1β levels, RIP1, RIP3, MLKL, NLRP3 and ASC expressions, col I and col III, TIMP4 expressions, the apoptosis rate and necrosis rate were lower than in HG group. Daidzin, the antagonist of ALDH2 abolished the role of Alda-1. In summary, ALDH2 maybe is a key regulator in high glucose induced cardiomyocytes injury. Activation of ALDH2 prevented the happening of fibrosis, apoptosis and necroptosis in high glucose induced primary cardiomyocytes injury model, the protective effects were related to the inhibiting of oxidative stress and inflammation, changing of MMP14 and TIMP4, then inhibiting the happening of fibrosis, apoptosis and necroptosis. These findings advance our understanding of the intensive mechanisms of ALDH2's cardioprotection, and provide the targeted basis for clinical diabetes treatment.
Collapse
Affiliation(s)
- Pinfang Kang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jiahui Wang
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Dian Fang
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Tingting Fang
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Ying Yu
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Weiping Zhang
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Lin Shen
- Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Zhenghong Li
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Hongju Wang
- Department of Cardiovascular Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233004, PR China
| | - Hongwei Ye
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| | - Qin Gao
- Department of Physiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Cardiovascular Research Center, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| |
Collapse
|
21
|
Elimination of Osteosarcoma by Necroptosis with Graphene Oxide-Associated Anti-HER2 Antibodies. Int J Mol Sci 2019; 20:ijms20184360. [PMID: 31491952 PMCID: PMC6770144 DOI: 10.3390/ijms20184360] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/15/2022] Open
Abstract
The prognosis for non-resectable or recurrent osteosarcoma (OS) remains poor. The finding that the majority of OS overexpress the protooncogene HER2 raises the possibility of using HER2 as a therapeutic target. However, clinical trials on the anti-HER2 antibody trastuzumab (TRA) in treating OS find no therapeutic benefit. HER2 overexpression in OS is not generally associated with gene amplification, with low-level expression regarded as HER2 “negative”, as per criteria used to classify breast cancer HER2 status. Nevertheless, active HER2-targeting approaches, such as virus-based HER2 vaccines or CAR-T cells have generated promising results. More recently, it has been found that the noncovalent association of TRA with nanomaterial graphene oxide (GO) generates stable TRA/GO complexes capable of rapidly killing OS cells. TRA/GO induces oxidative stress and strong HER2 signaling to elicit immediate degradation of both cIAP (cellular inhibitor of apoptosis protein) and caspase 8, leading to activation of necroptosis. This is an attractive mechanism of cancer cell death as chemo/apoptosis-resistant tumors may remain susceptible to necroptosis. In addition, necroptosis is potentially immunogenic to promote tumor immunity, as opposed to apoptosis that tends to silence tumor immunity. Currently, no established anticancer therapeutics are known to eliminate cancers by necroptosis. The aim of this article is to review the rationale and mechanisms of TRA/GO-mediated cytotoxicity.
Collapse
|