1
|
García-García CA, Cruz-Gregorio A, Pedraza-Chaverri J, Montaño LF, Rendón-Huerta EP. NDMA enhances claudin-1 and -6 expression viaCYP2E1/ROS in AGS cells. Toxicol In Vitro 2025; 102:105952. [PMID: 39395750 DOI: 10.1016/j.tiv.2024.105952] [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: 06/19/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 10/14/2024]
Abstract
Carcinogenic N-nitroso compounds, especially N-nitroso dimethylamine, increase the risk of gastric cancer development. Cytochrome P450-2E1 metabolizes this compound, thus generating an oxidant microenvironment. We aimed to evaluate in gastric adenocarcinoma cells if its effect on CYP2E1 and ROS affects signaling pathways associated with gastric cancer oncogenesis. The impact of N- nitroso dimethylamine upon CYP2E1 and ROS activation/secretion was evaluated by the DCFDA assay protocol, TER measurements, Stat3, pSTAT3, ERK1/2, and pERK1/2 expression, claudins-1 and -6 expression, and finally mRNA values of IL-1β IL-6, IL-8 and TNFα. Our results showed that exposure to N- N-nitroso dimethylamine disrupts the regulation of Stat3 and Erk1/2, alters the expression of claudin-1 and claudin-6 tight junction proteins, and increases the secretion of pro-inflammatory cytokines. These alterations induce a continuous local inflammatory process, an event identified as a gastric cancer promoter. In summary, N-nitroso dimethylamine can disrupt cell mechanisms associated with gastric cancer oncogenesis.
Collapse
Affiliation(s)
| | - Alfredo Cruz-Gregorio
- Departamento de Fisiología, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico
| | | | - Luis F Montaño
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico
| | - Erika P Rendón-Huerta
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Tisular, Facultad de Medicina, UNAM, Mexico.
| |
Collapse
|
2
|
Yun HM, Kim SH, Kwon YJ, Park KR. Effect of Spicatoside a on Anti-Osteosarcoma MG63 Cells through Reactive Oxygen Species Generation and the Inhibition of the PI3K-AKT-mTOR Pathway. Antioxidants (Basel) 2024; 13:1162. [PMID: 39456416 PMCID: PMC11505237 DOI: 10.3390/antiox13101162] [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: 08/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Osteosarcoma is a primary malignant tumor found in the bones of children and adolescents. Unfortunately, many patients do not respond well to treatment and succumb to the illness. Therefore, it is necessary to discover novel bioactive compounds to overcome therapeutic limitations. Liriope platyphylla Wang et Tang is a well-known herb used in oriental medicine. Studies have shown that metabolic diseases can be clinically treated using the roots of L. platyphylla. Recent studies have demonstrated the anticarcinoma potential of root extracts; however, the exact mechanism remains unclear. The aim of this study was to examine the anti-osteosarcoma activity of a single compound extracted from the dried roots of L. platyphylla. We purified Spicatoside A (SpiA) from the dried roots of L. platyphylla. SpiA significantly inhibited the proliferation of human osteosarcoma MG63 cells in a dose- and time-dependent manner. SpiA also regulated the expression of various downstream proteins that mediate apoptosis (PARP, Bcl-2, and Bax), cell growth (cyclin D1, Cdk4, and Cdk6), angiogenesis (VEGF), and metastasis (MMP13). The Proteome Profiler Human Phospho-Kinase Array Kit showed that the AKT signaling protein was a target of SpiA in osteosarcoma cells. We also found that SpiA suppressed the constitutive activation of the PI3K-AKT-mTOR-p70S6K1 signaling pathway. We further validated the effects of SpiA on the AKT signaling pathway. SpiA induced autophagosome formation and suppressed necroptosis (a form of programmed cell death). SpiA increased the generation of reactive oxygen species (ROS) and led to the loss of mitochondrial membrane potential. N-acetylcysteine (NAC)-induced inhibition of ROS generation reduced SpiA-induced AKT inhibition, apoptotic cell death, and anti-metastatic effects by suppressing cell migration and invasion. Overall, these results highlight the anti-osteosarcoma effect of SpiA by inhibiting the AKT signaling pathway through ROS generation, suggesting that SpiA may be a promising compound for the treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Soo Hyun Kim
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.-J.K.)
| | - Yoon-Ju Kwon
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.-J.K.)
| | - Kyung-Ran Park
- Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
3
|
Afonso MB, David JC, Alves MI, Santos AA, Campino G, Ratziu V, Gautheron J, Rodrigues CMP. Intricate interplay between cell metabolism and necroptosis regulation in metabolic dysfunction-associated steatotic liver disease: A narrative review. Metabolism 2024; 158:155975. [PMID: 39004396 DOI: 10.1016/j.metabol.2024.155975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), encompasses a progressive spectrum of liver conditions, ranging from steatosis to metabolic dysfunction-associated steatohepatitis, characterised by hepatocellular death and inflammation, potentially progressing to cirrhosis and/or liver cancer. In both experimental and human MASLD, necroptosis-a regulated immunogenic necrotic cell death pathway-is triggered, yet its exact role in disease pathogenesis remains unclear. Noteworthy, necroptosis-related signalling pathways are emerging as key players in metabolic reprogramming, including lipid and mitochondrial metabolism. Additionally, metabolic dysregulation is a well-established contributor to MASLD development and progression. This review explores the intricate interplay between cell metabolism and necroptosis regulation and its impact on MASLD pathogenesis. Understanding these cellular events may offer new insights into the complexity of MASLD pathophysiology, potentially uncovering therapeutic opportunities and unforeseen metabolic consequences of targeting necroptosis.
Collapse
Affiliation(s)
- Marta Bento Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Jan Caira David
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Mariana Isabel Alves
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - André Anastácio Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Gonçalo Campino
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Vlad Ratziu
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Department of Hepatology, Paris, France; Sorbonne Université, Inserm, Centre de Recherche des Cordeliers (CRC), Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Jérémie Gautheron
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, France; Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | | |
Collapse
|
4
|
Yun HM, Kwon HS, Lee JY, Park KR. Vitexicarpin Induces Apoptosis and Inhibits Metastatic Properties via the AKT-PRAS40 Pathway in Human Osteosarcoma. Int J Mol Sci 2024; 25:3582. [PMID: 38612399 PMCID: PMC11012096 DOI: 10.3390/ijms25073582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Osteosarcoma, which has poor prognosis after metastasis, is the most common type of bone cancer in children and adolescents. Therefore, plant-derived bioactive compounds are being actively developed for cancer therapy. Artemisia apiacea Hance ex Walp. is a traditional medicinal plant native to Eastern Asia, including China, Japan, and Korea. Vitexicarpin (Vitex), derived from A. apiacea, has demonstrated analgesic, anti-inflammatory, antitumour, and immunoregulatory properties; however, there are no published studies on Vitex isolated from the aerial parts of A. apiacea. Thus, this study aimed to evaluate the antitumour activity of Vitex against human osteosarcoma cells. In the present study, Vitex (>99% purity) isolated from A. apiacea induced significant cell death in human osteosarcoma MG63 cells in a dose- and time-dependent manner; cell death was mediated by apoptosis, as evidenced by the appearance of cleaved-PARP, cleaved-caspase 3, anti-apoptotic proteins (Survivin and Bcl-2), pro-apoptotic proteins (Bax), and cell cycle-related proteins (Cyclin D1, Cdk4, and Cdk6). Additionally, a human phosphokinase array proteome profiler revealed that Vitex suppressed AKT-dependent downstream kinases. Further, Vitex reduced the phosphorylation of PRAS40, which is associated with autophagy and metastasis, induced autophagosome formation, and suppressed programmed cell death and necroptosis. Furthermore, Vitex induced antimetastatic activity by suppressing the migration and invasion of MMP13, which is the primary protease that degrades type I collagen for tumour-induced osteolysis in bone tissues and preferential metastasis sites. Taken together, our results suggest that Vitex is an attractive target for treating human osteosarcoma.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Sook Kwon
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (H.S.K.); (J.Y.L.)
| | - Joon Yeop Lee
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (H.S.K.); (J.Y.L.)
| | - Kyung-Ran Park
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
5
|
Xie Y, Zhao G, Lei X, Cui N, Wang H. Advances in the regulatory mechanisms of mTOR in necroptosis. Front Immunol 2023; 14:1297408. [PMID: 38164133 PMCID: PMC10757967 DOI: 10.3389/fimmu.2023.1297408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Snyder K, Gorse K, Kochanek PM, Jackson TC. Neuronal RBM5 modulates cell signaling responses to traumatic and hypoxic-ischemic injury in a sex-dependent manner. Cell Death Discov 2023; 9:379. [PMID: 37848418 PMCID: PMC10582027 DOI: 10.1038/s41420-023-01677-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023] Open
Abstract
It is not clear if inhibiting the pro-death gene RNA binding motif 5 (RBM5) is neuroprotective in isolated primary neurons or if it regulates cell survival in a sex-dependent manner. Here we established sex-dichotomized primary cortical neuron cultures from transgenic mice harboring a floxed RBM5 gene-trap. Lentivirus-mediated expression of CRE was used to silence RBM5 expression. Male and female neurons were maintained in next-generation Neurobasal-Plus media and subjected to a mechanical stretch-injury (to model traumatic brain injury) or oxygen-glucose deprivation/OGD (to model ischemia). RBM5 KO did not affect 24 h post-injury survival as determined by lactate dehydrogenase (LDH) release, in either paradigm. In contrast, female KO neurons had increased spectrin breakdown products post-insult (in both models). Furthermore, in OGD, RBM5 KO in male neurons exacerbated injury-induced downregulation of pro-survival AKT activation (pAKT473) but conversely led to pAKT473 sparing in female neurons. Moreover, global proteomics identified 19 differentially expressed (DE) proteins in OGD-injured male neurons, and 102 DE proteins in injured female neurons. Two novel RBM5-regulated proteins (PIGQ and EST1C) were identified in injured male KO neurons, and 8 novel proteins identified in injured female KO neurons (S35A5, DHTK1, STX3, IF3M, RN167, K1C14, DYHS, and MED13). In summary, RBM5 inhibition does not modify neuronal survival in primary mouse neurons in 2 clinically relevant models of excitotoxic insult, but RBM5 does regulate intracellular responses to injury in a sex-dependent manner.
Collapse
Affiliation(s)
- Kara Snyder
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Kiersten Gorse
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Travis C Jackson
- University of South Florida, Morsani College of Medicine, USF Health Heart Institute, MDD 0630, 560 Channelside Dr, Tampa, FL, 33602, USA.
- University of South Florida, Morsani College of Medicine, Department of Molecular Pharmacology & Physiology, 12901 Bruce B Downs Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
7
|
Xu J, Li Y, Kang M, Chang C, Wei H, Zhang C, Chen Y. Multiple forms of cell death: A focus on the PI3K/AKT pathway. J Cell Physiol 2023; 238:2026-2038. [PMID: 37565518 DOI: 10.1002/jcp.31087] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023]
Abstract
Cell death is a natural biological process that occurs in living organisms. Since 1963, extensive research has shed light on the occurrence, progress, and final outcome of cell death. According to different cell phenotypes, it is classified into different types, including apoptosis, pyroptosis, necroptosis, autophagy, ferroptosis, cuproptosis, and so on. However, regardless of the form of cell death, what we ultimately expect is the disappearance of abnormal cells, such as tumor cells, while normal cells survive. As a result, it is vital to investigate the details of cell death, including death triggers, potent regulators, and executioners. Although significant progress has been made in understanding molecular pathways of cell death, many aspects remain unclear because of the complex regulatory networks in cells. Among them, the phosphoinositide-3-kinase (PI3K)/protein kinase B(AKT) pathway is discovered to be a crucial regulator of the cell death process. AKT, as a proto-oncogene, has become a major focus of attention in the medical community due to its role in regulating a multiplicity of cellular functions counting metabolism, immunity, proliferation, survival, transcription, and protein synthesis. Here, we explored the connection between the PI3K/AKT pathway and cell death, aiming to enhance our comprehension of the mechanism underlying this process. Such knowledge may pave the way for the subsequent development of more effective disease treatments, such as finding suitable targets for drug intervention.
Collapse
Affiliation(s)
- Jiawei Xu
- Department of Medical Science Research Center, Peihua University, Xi'an, Shaanxi, China
| | - Yu Li
- Department of Medical Science Research Center, Peihua University, Xi'an, Shaanxi, China
| | - Meili Kang
- Department of Medical Science Research Center, Peihua University, Xi'an, Shaanxi, China
| | - Cuicui Chang
- Department of Medical Science Research Center, Peihua University, Xi'an, Shaanxi, China
| | - Hong Wei
- Department of Rehabilitation Teaching and Research, Xi'an Siyuan University, Xi'an, China
| | - Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, China
| | - Yuhua Chen
- Department of Neurosurgery, Life Science Research Laboratory, Bijie Traditional Chinese Medicine Hospital, Bijie, China
| |
Collapse
|
8
|
Cheung J, Remiszewski S, Chiang LW, Ahmad E, Pal M, Rahman SA, Nikolovska-Coleska Z, Chan GC. Inhibition of SIRT2 promotes death of human cytomegalovirus-infected peripheral blood monocytes via apoptosis and necroptosis. Antiviral Res 2023; 217:105698. [PMID: 37562606 DOI: 10.1016/j.antiviral.2023.105698] [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: 05/24/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Peripheral blood monocytes are the cells predominantly responsible for systemic dissemination of human cytomegalovirus (HCMV) and a significant cause of morbidity and mortality in immunocompromised patients. HCMV establishes a silent/quiescent infection in monocytes, which is defined by the lack of viral replication and lytic gene expression. The absence of replication shields the virus within infected monocytes from the current available antiviral drugs that are designed to suppress active replication. Our previous work has shown that HCMV stimulates a noncanonical phosphorylation of Akt and the subsequent upregulation of a distinct subset of prosurvival proteins in normally short-lived monocytes. In this study, we found that SIRT2 activity is required for the unique activation profile of Akt induced within HCMV-infected monocytes. Importantly, both therapeutic and prophylactic treatment with a novel SIRT2 inhibitor, FLS-379, promoted death of infected monocytes via both the apoptotic and necroptotic cell death pathways. Mechanistically, SIRT2 inhibition reduced expression of Mcl-1, an Akt-dependent antiapoptotic Bcl-2 family member, and enhanced activation of MLKL, the executioner kinase of necroptosis. We have previously reported HCMV to block necroptosis by stimulating cellular autophagy. Here, we additionally demonstrate that inhibition of SIRT2 suppressed Akt-dependent HCMV-induced autophagy leading to necroptosis of infected monocytes. Overall, our data show that SIRT2 inhibition can simultaneously promote death of quiescently infected monocytes by two distinct death pathways, apoptosis and necroptosis, which may be vital for limiting viral dissemination to peripheral organs in immunosuppressed patients.
Collapse
Affiliation(s)
- Jennifer Cheung
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Stacy Remiszewski
- Evrys Bio, LLC, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA
| | - Lillian W Chiang
- Evrys Bio, LLC, Pennsylvania Biotechnology Center, Doylestown, PA, 18902, USA
| | - Ejaz Ahmad
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Mohan Pal
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Sm Ashikur Rahman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Zaneta Nikolovska-Coleska
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gary C Chan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
9
|
Wang Y, Hong Y, Mao S, Pan J, Cui Y, Lu J, Wen T, Wang X, Luo Y. Downregulation of miR-124-3p suppresses the development of the deep retinal blood vessels by enhancing the Stat1/Ripk1 pathway in mouse retinal microglia. Exp Eye Res 2023; 233:109551. [PMID: 37356537 DOI: 10.1016/j.exer.2023.109551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 06/18/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
The study aimed to investigate the role of microRNA (miR)-124-3p in retinal angiogenesis in a mouse model. An intravitreal injection of miR-124-3p antagomir was used to knockdown the expression of miR-124-3p in the mouse retina at postnatal day (P)3. Immunofluorescent staining of both retinal frozen sections and whole retina were used to observe retinal vascular development in the P6, P9 and P12 mice, as well as the changes in retinal ganglion cells, astrocytes, Müller cells and microglia. Whole retinal RNA extracted from P9 mice was used for transcriptome sequencing. Following gene set enrichment analysis, the enriched genes caused by miR-124-3p inhibition were analyzed by immunofluorescent staining and western blot. Results indicated that deep vascular development was significantly inhibited by the activation of M1 phenotype microglia. Moreover, there were no notable effects on superficial retinal vascular development, the retinal ganglion cells, astrocytes, and Müller cells. The expression of the Stat1/Irf9/Eif2ak2/Ripk1 axis in the miR-124-3p knockdown group was significantly increased. The microglia penetrated deep into the retina and the activation of Ripk1(+) microglia significantly increased, which was accompanied by an increased level of apoptosis to inhibit the deep vascular sprout. Downregulation of miR-124-3p during the early retinal development can suppress the development of the deep retinal blood vessels by enhancing the expression level of the Stat1/Irf9/Eif2ak2/Ripk1 axis and inducing the cell apoptosis of the activation of Ripk1(+) microglia.
Collapse
Affiliation(s)
- Yishen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yiwen Hong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Shudi Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yamei Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jing Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Tao Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Wang H, Zheng A, Arias EB, Kwak SE, Pan X, Duan D, Cartee GD. AS160 expression, but not AS160 Serine-588, Threonine-642, and Serine-704 phosphorylation, is essential for elevated insulin-stimulated glucose uptake by skeletal muscle from female rats after acute exercise. FASEB J 2023; 37:e23021. [PMID: 37289137 DOI: 10.1096/fj.202300282rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/09/2023]
Abstract
One exercise session can increase subsequent insulin-stimulated glucose uptake (ISGU) by skeletal muscle in both sexes. We recently found that muscle expression and phosphorylation of key sites of Akt substrate of 160 kDa (AS160; also called TBC1D4) are essential for the full-exercise effect on postexercise-ISGU (PEX-ISGU) in male rats. In striking contrast, AS160's role in increased PEX-ISGU has not been rigorously tested in females. Our rationale was to address this major knowledge gap. Wild-type (WT) and AS160-knockout (KO) rats were either sedentary or acutely exercised. Adeno-associated virus (AAV) vectors were engineered to express either WT-AS160 or AS160 mutated on key serine and threonine residues (Ser588, Thr642, and Ser704) to alanine to prevent their phosphorylation. AAV vectors were delivered to the muscle of AS160-KO rats to determine if WT-AS160 or phosphorylation-inactivated AS160 would influence PEX-ISGU. AS160-KO rats have lower skeletal muscle abundance of the GLUT4 glucose transporter protein. This GLUT4 deficit was rescued using AAV delivery of GLUT4 to determine if eliminating muscle GLUT4 deficiency would normalize PEX-ISGU. The novel results were as follows: (1) AS160 expression was required for greater PEX-ISGU; (2) rescuing muscle AS160 expression in AS160-KO rats restored elevated PEX-ISGU; (3) AS160's essential role for the postexercise increase in ISGU was not attributable to reduced muscle GLUT4 content; and (4) AS160 phosphorylation on Ser588, Thr642, and Ser704 was not essential for greater PEX-ISGU. In conclusion, these novel findings revealed that three phosphosites widely proposed to influence PEX-ISGU are not required for this important outcome in female rats.
Collapse
Affiliation(s)
- Haiyan Wang
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy Zheng
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Edward B Arias
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Seong Eun Kwak
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiufang Pan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, USA
| | - Gregory D Cartee
- Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Xie Y, Lei X, Zhao G, Guo R, Cui N. mTOR in programmed cell death and its therapeutic implications. Cytokine Growth Factor Rev 2023; 71-72:66-81. [PMID: 37380596 DOI: 10.1016/j.cytogfr.2023.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023]
Abstract
Mechanistic target of rapamycin (mTOR), a highly conserved serine/threonine kinase, is involved in cellular metabolism, protein synthesis, and cell death. Programmed cell death (PCD) assists in eliminating aging, damaged, or neoplastic cells, and is indispensable for sustaining normal growth, fighting pathogenic microorganisms, and maintaining body homeostasis. mTOR has crucial functions in the intricate signaling pathway network of multiple forms of PCD. mTOR can inhibit autophagy, which is part of PCD regulation. Cell survival is affected by mTOR through autophagy to control reactive oxygen species production and the degradation of pertinent proteins. Additionally, mTOR can regulate PCD in an autophagy-independent manner by affecting the expression levels of related genes and phosphorylating proteins. Therefore, mTOR acts through both autophagy-dependent and -independent pathways to regulate PCD. It is conceivable that mTOR exerts bidirectional regulation of PCD, such as ferroptosis, according to the complexity of signaling pathway networks, but the underlying mechanisms have not been fully explained. This review summarizes the recent advances in understanding mTOR-mediated regulatory mechanisms in PCD. Rigorous investigations into PCD-related signaling pathways have provided prospective therapeutic targets that may be clinically beneficial for treating various diseases.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ran Guo
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
12
|
Gupta R, Kumari S, Tripathi R, Ambasta RK, Kumar P. Unwinding the modalities of necrosome activation and necroptosis machinery in neurological diseases. Ageing Res Rev 2023; 86:101855. [PMID: 36681250 DOI: 10.1016/j.arr.2023.101855] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/09/2022] [Accepted: 01/15/2023] [Indexed: 01/20/2023]
Abstract
Necroptosis, a regulated form of cell death, is involved in the genesis and development of various life-threatening diseases, including cancer, neurological disorders, cardiac myopathy, and diabetes. Necroptosis initiates with the formation and activation of a necrosome complex, which consists of RIPK1, RIPK2, RIPK3, and MLKL. Emerging studies has demonstrated the regulation of the necroptosis cell death pathway through the implication of numerous post-translational modifications, namely ubiquitination, acetylation, methylation, SUMOylation, hydroxylation, and others. In addition, the negative regulation of the necroptosis pathway has been shown to interfere with brain homeostasis through the regulation of axonal degeneration, mitochondrial dynamics, lysosomal defects, and inflammatory response. Necroptosis is controlled by the activity and expression of signaling molecules, namely VEGF/VEGFR, PI3K/Akt/GSK-3β, c-Jun N-terminal kinases (JNK), ERK/MAPK, and Wnt/β-catenin. Herein, we briefly discussed the implication and potential of necrosome activation in the pathogenesis and progression of neurological manifestations, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, traumatic brain injury, and others. Further, we present a detailed picture of natural compounds, micro-RNAs, and chemical compounds as therapeutic agents for treating neurological manifestations.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), India.
| |
Collapse
|
13
|
Xu W, Yang T, Lou X, Chen J, Wang X, Hu M, An D, Gao R, Wang J, Chen X. Role of the Peli1-RIPK1 Signaling Axis in Methamphetamine-Induced Neuroinflammation. ACS Chem Neurosci 2023; 14:864-874. [PMID: 36763609 DOI: 10.1021/acschemneuro.2c00623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
Abstract
Severe neurological inflammation is one of the main symptoms of methamphetamine (meth)-induced brain injury. Studies have demonstrated that meth exposure facilitates neuroinflammation via Pellino E3 ubiquitin protein ligase 1 (Peli1)-mediated signaling. However, the involved mechanisms remain incompletely understood. Herein, we used Peli1-/- mice and Peli1-knockdown microglial BV2 cells to decipher the roles of Peli1 and downstream signaling in meth-induced neuroinflammation. After meth administration for seven consecutive days, Peli1-/- mice exhibited better learning and memory behavior and dramatically lower interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-6 levels than wild-type mice. Moreover, in vitro experiments revealed that Peli1 knockdown significantly attenuated the meth-induced upregulation of cytokines. Besides, meth markedly activated and increased the levels of receptor-interacting protein kinase 1 (RIPK1), and Peli1 knockout or knockdown prevented these effects, indicating that RIPK1 participated in meth-induced Peli1-mediated inflammation. Specifically, treating the cells with necrostatin-1(Nec-1), an antagonist of RIPK1, remarkably inhibited the meth-induced increase in IL-1β, TNF-α, and IL-6 expression, confirming the involvement of RIPK1 in Peli1-mediated neuroinflammation. Finally, meth induced a dramatic transfer of the mixed lineage kinase domain-like protein, a downstream effector of RIRK1, to the cell membrane, disrupting membrane integrity and causing cytokine excretion. Therefore, targeting the Peli1-RIPK1 signaling axis is a potentially valid therapeutic approach against meth-induced neuroinflammation.
Collapse
Affiliation(s)
- Weixiao Xu
- Department of Emergency Medicine, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Tingyu Yang
- Wujin District Center for Disease Prevention and Control, Changzhou 213100, Jiangsu, China
| | - Xinyu Lou
- The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, Jiangsu, China
| | - Jingrong Chen
- The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, Jiangsu, China
| | - Xi Wang
- The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, Jiangsu, China
| | - Miaoyang Hu
- The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, Jiangsu, China
| | - Di An
- Department of Emergency Medicine, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| | - Rong Gao
- Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jun Wang
- The Key Lab of Modern Toxicology (NJMU), Ministry of Education, School of Public Health, Nanjing Medical University, 818 Tianyuan East Road, Nanjing 211166, Jiangsu, China
| | - Xufeng Chen
- Department of Emergency Medicine, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, 300 Guangzhou Road, Nanjing 210029, Jiangsu, China
| |
Collapse
|
14
|
Naeem A, Knoer G, Avantaggiati ML, Rodriguez O, Albanese C. Provocative non-canonical roles of p53 and AKT signaling: A role for Thymosin β4 in medulloblastoma. Int Immunopharmacol 2023; 116:109785. [PMID: 36720193 DOI: 10.1016/j.intimp.2023.109785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2023]
Abstract
The PI3K/AKT and p53 pathways are key regulators of cancer cell survival and death, respectively. Contrary to their generally accepted roles, several lines of evidence, including ours in medulloblastoma, the most common childhood brain cancer, highlight non-canonical functions for both proteins and show a complex context-dependent dynamic behavior in determining cell fate. Interestingly, p53-mediated cell survival and AKT-mediated cell death can dominate in certain conditions, and these interchangeable physiological functions may potentially be manipulated for better clinical outcomes. This review article presents studies in which p53 and AKT behave contrary to their well-established functions. We discuss the factors and circumstances that may be involved in mediating these changes and the implications of these unique roles of p53 and AKT in devising therapeutic strategies. Lastly, based on our recent finding of Thymosin beta 4-mediated chemosensitivity via an AKT-p53 interaction in medulloblastoma cells, we also discuss the possible implications of Thymosin beta-4 in enhancing drug sensitivity in this deadly childhood disease.
Collapse
Affiliation(s)
- Aisha Naeem
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Health Research Governance Department, Ministry of Public Health, Qatar.
| | - Grace Knoer
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Maria Laura Avantaggiati
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Olga Rodriguez
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chris Albanese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; Department of Radiology, Georgetown University Medical Center, Washington, DC 20057, USA; Center for Translational Imaging, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
15
|
Chaouhan HS, Vinod C, Mahapatra N, Yu SH, Wang IK, Chen KB, Yu TM, Li CY. Necroptosis: A Pathogenic Negotiator in Human Diseases. Int J Mol Sci 2022; 23:12714. [PMID: 36361505 PMCID: PMC9655262 DOI: 10.3390/ijms232112714] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
Over the past few decades, mechanisms of programmed cell death have attracted the scientific community because they are involved in diverse human diseases. Initially, apoptosis was considered as a crucial mechanistic pathway for programmed cell death; recently, an alternative regulated mode of cell death was identified, mimicking the features of both apoptosis and necrosis. Several lines of evidence have revealed that dysregulation of necroptosis leads to pathological diseases such as cancer, cardiovascular, lung, renal, hepatic, neurodegenerative, and inflammatory diseases. Regulated forms of necrosis are executed by death receptor ligands through the activation of receptor-interacting protein kinase (RIPK)-1/3 and mixed-lineage kinase domain-like (MLKL), resulting in the formation of a necrosome complex. Many papers based on genetic and pharmacological studies have shown that RIPKs and MLKL are the key regulatory effectors during the progression of multiple pathological diseases. This review focused on illuminating the mechanisms underlying necroptosis, the functions of necroptosis-associated proteins, and their influences on disease progression. We also discuss numerous natural and chemical compounds and novel targeted therapies that elicit beneficial roles of necroptotic cell death in malignant cells to bypass apoptosis and drug resistance and to provide suggestions for further research in this field.
Collapse
Affiliation(s)
- Hitesh Singh Chaouhan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Ch Vinod
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Nikita Mahapatra
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Shao-Hua Yu
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - I-Kuan Wang
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| | - Tung-Min Yu
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40402, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| |
Collapse
|
16
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
17
|
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]
|
18
|
Dan W, Zhong L, Zhang Z, Wan P, Lu Y, Wang X, Liu Z, Chu X, Liu B. RIP1-dependent Apoptosis and Differentiation Regulated by Skp2 and Akt/GSK3β in Acute Myeloid Leukemia. Int J Med Sci 2022; 19:525-536. [PMID: 35370472 PMCID: PMC8964317 DOI: 10.7150/ijms.68385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 02/22/2022] [Indexed: 11/05/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous neoplasm characterized by variations in cytogenetics and molecular abnormalities, which result in variable response to therapy. Receptor-interacting serine/threonine kinase 1 (RIP1)-mediated necroptosis has been reported to have a potential role in the treatment of AML. We obtained Skp2 and RIP1 are significantly overexpressed in AML samples using original published data, and identified that Skp2-depletion in AML cells significantly suppressed RIP1. Functional analysis showed that the inhibition of RIP1 caused by necrostatin-1 (Nec-1) inhibited the proliferation, simultaneously facilitate both the apoptosis and differentiation of AML cells. Mechanistical analysis elucidated that knockdown of Skp2 suppresses RIP1 by transcriptional regulation but not by proteasome degradation. Additionally, Skp2 regulated the function of RIP1 by decreasing K63-linked ubiquitin interaction with RIP1. Moreover, the suppression of Akt/GSK3β was observed in Skp2 knockdown stable NB4 cells. Also, GSK3β inactivation via small-molecule inhibitor treatment remarkably decreased RIP1 level. RIP1 regulates differentiation by interacting with RARα, increasing RA signaling targets gene C/EBPα and C/EBPβ. In conclusion, our study provides a novel insight into the mechanism of tumorigenesis and the development of AML, for which the Skp2-Akt/GSK3β-RIP1 pathway can be developed as a promising therapeutic target.
Collapse
Affiliation(s)
- Wenran Dan
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China.,Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zhonghui Zhang
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Peng Wan
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China.,Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yang Lu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xiao Wang
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhenyan Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xuan Chu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China.,Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Beizhong Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China.,Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
19
|
Wang H, Arias EB, Treebak JT, Cartee GD. Exercise effects on γ3-AMPK activity, Akt substrate of 160 kDa phosphorylation, and glucose uptake in muscle of normal and insulin-resistant female rats. J Appl Physiol (1985) 2022; 132:140-153. [PMID: 34882030 PMCID: PMC8759959 DOI: 10.1152/japplphysiol.00533.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Previous studies demonstrated that acute exercise can enhance glucose uptake (GU), γ3-AMP-activated protein kinase (AMPK) activity, and Akt substrate of 160 kDa (AS160) phosphorylation in skeletal muscles from low-fat diet (LFD)- and high-fat diet (HFD)-fed male rats. Because little is known about exercise effects on these outcomes in females, we assessed postexercise GU by muscles incubated ± insulin, delta-insulin GU (GU of muscles incubated with insulin minus GU uptake of paired muscles incubated without insulin), and muscle signaling proteins from female rats fed a LFD or a brief HFD (2 wk). Rats were sedentary (LFD-SED, HFD-SED) or swim exercised. Immediately postexercise (IPEX) or 3 h postexercise (3hPEX), epitrochlearis muscles were incubated (no insulin IPEX; ±insulin 3hPEX) to determine GU. Muscle γ3-AMPK activity (IPEX, 3hPEX) and phosphorylated AS160 (pAS160; 3hPEX) were also assessed. γ3-AMPK activity and insulin-independent GU of IPEX rats exceeded sedentary rats without diet-related differences in either outcome. At 3hPEX, both GU by insulin-stimulated muscles and delta-insulin GU exceeded their respective diet-matched sedentary controls. GU by insulin-stimulated muscles, but not delta-insulin GU for LFD-3hPEX, exceeded HFD-3hPEX. LFD-3hPEX versus LFD-SED had greater γ3-AMPK activity and greater pAS160. HFD-3hPEX exceeded HFD-SED for pAS160 but not for γ3-AMPK activity. pAS160 and γ3-AMPK at 3hPEX did not differ between diet groups. These results revealed that increased γ3-AMPK activity at 3hPEX was not essential for greater GU in insulin-stimulated muscle or greater delta-insulin GU in HFD female rats. Similarly elevated γ3-AMPK activity in LFD-IPEX versus HFD-IPEX and pAS160 in LFD-3hPEX versus HFD-3hPEX may contribute to the comparable delta-insulin GU at 3hPEX in both diet groups.NEW & NOTEWORTHY Glucose uptake (GU) and phosphorylated AS160 (pAS160) by insulin-stimulated muscles at 3 h postexercise (3hPEX) exceeded diet-matched controls in female low-fat diet-fed (LFD) or high-fat diet-fed (HFD) rats. GU with insulin for LFD-3hPEX exceeded HFD-3hPEX, whereas pAS160 was similar between these groups. γ3-AMPK immediately postexercise (IPEX) was similarly elevated in LFD and HFD, but only LFD-3hPEX had increased γ3-AMPK. These results suggest that greater γ3-AMPK at IPEX and pAS160 at 3hPEX may contribute to elevated GU with insulin, but greater γ3-AMPK at 3hPEX was dispensable for female HFD rats.
Collapse
Affiliation(s)
- Haiyan Wang
- 1Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan
| | - Edward B. Arias
- 1Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan
| | - Jonas T. Treebak
- 2Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gregory D. Cartee
- 1Muscle Biology Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan,3Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan,4Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
20
|
Gupta R, Ambasta RK, Pravir Kumar. Autophagy and apoptosis cascade: which is more prominent in neuronal death? Cell Mol Life Sci 2021; 78:8001-8047. [PMID: 34741624 PMCID: PMC11072037 DOI: 10.1007/s00018-021-04004-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/16/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis are two crucial self-destructive processes that maintain cellular homeostasis, which are characterized by their morphology and regulated through signal transduction mechanisms. These pathways determine the fate of cellular organelle and protein involved in human health and disease such as neurodegeneration, cancer, and cardiovascular disease. Cell death pathways share common molecular mechanisms, such as mitochondrial dysfunction, oxidative stress, calcium ion concentration, reactive oxygen species, and endoplasmic reticulum stress. Some key signaling molecules such as p53 and VEGF mediated angiogenic pathway exhibit cellular and molecular responses resulting in the triggering of apoptotic and autophagic pathways. Herein, based on previous studies, we describe the intricate relation between cell death pathways through their common genes and the role of various stress-causing agents. Further, extensive research on autophagy and apoptotic machinery excavates the implementation of selective biomarkers, for instance, mTOR, Bcl-2, BH3 family members, caspases, AMPK, PI3K/Akt/GSK3β, and p38/JNK/MAPK, in the pathogenesis and progression of neurodegenerative diseases. This molecular phenomenon will lead to the discovery of possible therapeutic biomolecules as a pharmacological intervention that are involved in the modulation of apoptosis and autophagy pathways. Moreover, we describe the potential role of micro-RNAs, long non-coding RNAs, and biomolecules as therapeutic agents that regulate cell death machinery to treat neurodegenerative diseases. Mounting evidence demonstrated that under stress conditions, such as calcium efflux, endoplasmic reticulum stress, the ubiquitin-proteasome system, and oxidative stress intermediate molecules, namely p53 and VEGF, activate and cause cell death. Further, activation of p53 and VEGF cause alteration in gene expression and dysregulated signaling pathways through the involvement of signaling molecules, namely mTOR, Bcl-2, BH3, AMPK, MAPK, JNK, and PI3K/Akt, and caspases. Alteration in gene expression and signaling cascades cause neurotoxicity and misfolded protein aggregates, which are characteristics features of neurodegenerative diseases. Excessive neurotoxicity and misfolded protein aggregates lead to neuronal cell death by activating death pathways like autophagy and apoptosis. However, autophagy has a dual role in the apoptosis pathways, i.e., activation and inhibition of the apoptosis signaling. Further, micro-RNAs and LncRNAs act as pharmacological regulators of autophagy and apoptosis cascade, whereas, natural compounds and chemical compounds act as pharmacological inhibitors that rescue neuronal cell death through inhibition of apoptosis and autophagic cell death.
Collapse
Affiliation(s)
- Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Mechanical Engineering Building, Delhi Technological University (Formerly Delhi College of Engineering), Room# FW4TF3, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
- , Delhi, India.
| |
Collapse
|
21
|
Bi A, Guo Z, Yang G, Huang Y, Yin Z, Luo L. γ-glutamylcysteine suppresses cadmium-induced apoptosis in PC12 cells via regulating oxidative stress. Toxicology 2021; 465:153029. [PMID: 34767868 DOI: 10.1016/j.tox.2021.153029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/06/2021] [Accepted: 11/06/2021] [Indexed: 02/07/2023]
Abstract
Cadmium (Cd) is a highly toxic environmental pollutant, leading to the occurrence and development of multiple neurological diseases. γ-glutamylcysteine (γ-GC) is a dipeptide formed by the condensation of l-glutamic acid and l-cysteine, which has antioxidant, anti-inflammatory, and chelating properties. The purpose of this study is to investigate the effect of γ-GC on Cd-induced apoptosis in PC12 cells. PC12 cells were pretreated with or without γ-GC (2 mM or 4 mM) for 2 h and exposed to Cd (10 μM) for 12 h, and survival, apoptosis, and oxidative stress of PC12 cells were detected after different treatments. The results showed that γ-GC significantly inhibited cell viability reduction, apoptosis, and depolarization of mitochondrial transmembrane potential in Cd-treated PC12 cells, as indicated by CCK-8 assay, flow cytometry, TUNEL staining, and JC-1 detection. Western blot showed that γ-GC down-regulated the ratio of Bax/Bcl-2 and the protein levels of cytosolic cytopigment c, cleaved-caspase-9, cleaved-caspase-3, and cleaved-PARP. Mechanistically, γ-GC suppressed Cd-induced ROS production, MDA accumulation, and GSH depletion, and increased the activity of antioxidant enzymes. Cd-induced activation of MAPK and PI3K/Akt signaling pathways were inhibited by γ-GC treatment, while sustained phosphorylation of JNK, p38, or Akt reversed anti-apoptotic effects of γ-GC. These results suggested that γ-GC inhibited Cd-induced apoptosis in PC12 cells through decreasing oxidative stress and inhibiting the activation of MAPK and PI3K/Akt signaling pathways. γ-GC could be used as a potential protective agent against Cd neurotoxicity.
Collapse
Affiliation(s)
- Aijing Bi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Zhen Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Guocui Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Youfang Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, Jiangsu, China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
22
|
Ehigie AF, Wei P, Wei T, Yan X, Olorunsogo OO, Ojeniyi FD, Ehigie LO. Momordica charantia L. induces non-apoptotic cell death in human MDA-MB-436 breast and A549 lung cancer cells by disrupting energy metabolism and exacerbating reactive oxygen species' generation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114036. [PMID: 33753145 DOI: 10.1016/j.jep.2021.114036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/03/2021] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bitter melon, Momordica charantia L. (MC), is an ethnomedicinal plant cultivated in different climes. It's cytotoxic effect on several cancer cell lines has been evaluated. However, there have been contrasting reports on the actual mechanism (s) involved in the observed cell death induced by MC. AIMS OF THE STUDY To probe the mechanism of cell death induction in MDA-MB-436 (Breast) and A549 (lung) cancer cell lines treated with fractions (ethyl acetate, dichloromethane and hexane) derived from the aqueous extract of MC. MATERIALS AND METHODS Aqeous extract of the leaves of MC were fractionated using solvents of different polarities (ethyl acetate (D3), n-hexane (D4), dichloromethane (D5)). The cells were incubated with 100 and 125 μg/mL of the fractions 24 hours. Combination of fluorescence microscopy, enzyme assays, Western blot analyses and flow cytometry were employed in the study. RESULTS Treatment of the cells with MC fractions reduced Mitochondrial Membrane Potential (MMP) and intracellular ATP levels, while increasing reactive oxygen species levels without classical biochemical and morphological apoptotic features were seen. However, the fractions failed in upregulating either caspase-3 activation or cytochrome c release in the cancer cells. CONCLUSION Overall, these results suggest that the cytotoxic effect of MC on the selected cancer cells is mediated by loss of mitochondrial function via loss of respiration leading to cell death rather than by the classical release of cytochrome c or caspase-3 activated apoptosis.
Collapse
Affiliation(s)
- Adeola Folasade Ehigie
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Peng Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Taotao Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Olufunso O Olorunsogo
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | | | - Leonard Ona Ehigie
- Laboratories for Biomembrane Research and Biotechnology, Biochemistry Department, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
23
|
Wang J, Zhai Y, Ou M, Bian Y, Tang C, Zhang W, Cheng Y, Li G. Protective Effect of Lemon Peel Extract on Oxidative Stress in H9c2 Rat Heart Cell Injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2047-2058. [PMID: 34017169 PMCID: PMC8131012 DOI: 10.2147/dddt.s304624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/21/2021] [Indexed: 01/27/2023]
Abstract
Aim Lemon peel, a traditional Chinese medicine, was tested in this study for its novel application in inhibiting cellular oxidative stress, and the effect of lemon peel extract (LPE) on protecting H9c2 rat heart cells from oxidative stress was investigated. Methods The scavenging effects of LPE on 1,1-diphenyl-2-picryhydrazyl (DPPH) and 2,2’-azino-bis (3-ethylbenzthiazoline-6-sulfonic acid) (ABTS) free radicals were measured in extracellular experiments. The 3-(4,5-dimethyl-2-thiazolinyl)-2,5-diphenyl-2-h-tetrazolylammonium bromide (MTT) assay was used to detect the cell survival rate. The cell supernatant and intracellular oxidation-related indicators were detected by a kit, and the mRNA expression in H9c2 cells was detected by quantitative polymerase chain reaction (qPCR). The chemical substances of LPE were analyzed by high-performance liquid chromatography (HPLC). Results The results showed that LPE exhibited better DPPH and ABTS free radical scavenging abilities than vitamin C. Compared with the cells in the normal state (control group), the cell survival rate in the model group decreased, and the level of lactate dehydrogenase (LDH) increased, the levels of superoxide dismutase (SOD), catalase (CAT), and glutathione (GSH) decreased, and the content of malondialdehyde (MDA) increased. Compared with the control group, the expression of Bcl-2-related X protein (Bax), caspase-3, nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in the model group was increased, and the expression of B-cell lymphoma-2 (Bcl-2) was reduced. Compared with the model group, LPE treatment improved the cell survival rate, reduced the levels of LDH and MDA, increased the levels of SOD, CAT, and GSH, downregulated the expression of Bax, caspase-3, Nrf2 and HO-1, and upregulated the expression of Bcl-2. The composition analysis showed that LPE contained catechin, rutin, naringin, quercetin, and hesperidin. Conclusion The results indicated that LPE could protect H9c2 cells from oxidative stress through five active components. LPE has the potential to be developed into natural medicine or health food for the inhibition of cell oxidative damage.
Collapse
Affiliation(s)
- Jun Wang
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| | - Yulin Zhai
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China
| | - Mingguang Ou
- Guang'an Nongfeng Agricultural Development Co., Ltd, Sichuan, People's Republic of China
| | - Yunfeng Bian
- Guang'an Zheng Wang Agriculture Co., Ltd, Sichuan, People's Republic of China
| | - Chenglong Tang
- Yuanyang Hongtu Grapefruit Agricultural Technology Development Co., Ltd, Yunnan, People's Republic of China
| | - Wanchao Zhang
- Chongqing Institute of Medicinal Plant Cultivation, Chongqing, People's Republic of China.,National Patent Navigation Project (Chongqing) Research and Promotion Center, Chongqing, People's Republic of China.,Bio-Resource Research and Utilization Joint Key Laboratory of Sichuan and Chongqing, Chongqing, People's Republic of China
| | - Yujiao Cheng
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| | - Guijie Li
- Citrus Research Institute, Southwest University, Chongqing, People's Republic of China.,National Citrus Engineering Research Center, Chongqing, People's Republic of China
| |
Collapse
|
24
|
Zhang Z, Ju F, Chen F, Wu H, Chen J, Zhong J, Shao L, Zheng S, Wang L, Xue M. GDC-0326 Enhances the Effects of 5-Fu in Colorectal Cancer Cells by Inducing Necroptotic Death. Onco Targets Ther 2021; 14:2519-2530. [PMID: 33880032 PMCID: PMC8053532 DOI: 10.2147/ott.s302334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
AIM Chemoresistance to 5-fluorouracil (5-Fu) is common in colorectal cancer (CRC). Programmed necrosis (necroptosis) is an alternative form of programmed cell death regulated by receptor-interacting protein kinase (RIPK) 1 and 3, assumed as a novel target of cancer therapy. In this study, we aimed to explore whether a novel small molecular agent GDC-0326 could facilitate the effect of 5-Fu through necroptosis. MAIN METHODS Cell Counting Kit-8 (CCK-8) assay and colony formation were performed to confirm the function of GDC-0326 in CRC cells. Western blot and immunofluorescence were conducted to measure the altered expressions of RIPK1/RIPK3 induced by GDC-0326. Subcutaneous tumor models were used to evaluate the chemotherapeutic effects and concomitant side effects of GDC-0326 in vivo. KEY FINDINGS We found that GDC-0326 effectively suppressed the growth of CRC cells in a dose-dependent manner. The induction of necroptosis by GDC-0326 was correlated with the modulation of RIPK1 and RIPK3. Necrostatin-1 and GSK-872, inhibitors of RIPK1 and RIPK3, respectively, could rescue the cell death induced by GDC-0326. In addition, in vitro and in vivo studies showed that 5-Fu plus GDC-0326 evinced a better antitumor efficacy by suppressing tumor growth and increasing tumor necrosis with no increased toxicity. SIGNIFICANCE This study demonstrates that GDC-0326 plus 5-Fu has augmented antitumor efficacy and acceptable safety, which might be a promising therapeutic strategy for CRC patients in the future.
Collapse
Affiliation(s)
- Zizhen Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Fangyu Ju
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Fei Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Haoyue Wu
- Institute of Genetics and Department of Genetics, Zhejiang University School of Medicine, Hangzhou, 310012, People’s Republic of China
| | - Jingyu Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Jing Zhong
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Liming Shao
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Sheng Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Liangjing Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| | - Meng Xue
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310020, People’s Republic of China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310000, People’s Republic of China
- Zhejiang University Cancer Center, Hangzhou, 310000, People’s Republic of China
| |
Collapse
|
25
|
Ko KW, Milbrandt J, DiAntonio A. SARM1 acts downstream of neuroinflammatory and necroptotic signaling to induce axon degeneration. J Cell Biol 2021; 219:151915. [PMID: 32609299 PMCID: PMC7401797 DOI: 10.1083/jcb.201912047] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/08/2020] [Accepted: 04/27/2020] [Indexed: 12/29/2022] Open
Abstract
Neuroinflammation and necroptosis are major contributors to neurodegenerative disease, and axon dysfunction and degeneration is often an initiating event. SARM1 is the central executioner of pathological axon degeneration. Here, we demonstrate functional and mechanistic links among these three pro-degenerative processes. In a neuroinflammatory model of glaucoma, TNF-α induces SARM1-dependent axon degeneration, oligodendrocyte loss, and subsequent retinal ganglion cell death. TNF-α also triggers SARM1-dependent axon degeneration in sensory neurons via a noncanonical necroptotic signaling mechanism. MLKL is the final executioner of canonical necroptosis; however, in axonal necroptosis, MLKL does not directly trigger degeneration. Instead, MLKL induces loss of the axon survival factors NMNAT2 and STMN2 to activate SARM1 NADase activity, which leads to calcium influx and axon degeneration. Hence, these findings define a specialized form of axonal necroptosis. The demonstration that neuroinflammatory signals and necroptosis can act locally in the axon to stimulate SARM1-dependent axon degeneration identifies a therapeutically targetable mechanism by which neuroinflammation can stimulate axon loss in neurodegenerative disease.
Collapse
Affiliation(s)
- Kwang Woo Ko
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO
| | - Aaron DiAntonio
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO.,Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
26
|
Xin C, Guangliang S, Qing Z, Qingqing L, Hang Y, Yiming Z, Shu L. Astilbin protects chicken peripheral blood lymphocytes from cadmium-induced necroptosis via oxidative stress and the PI3K/Akt pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 190:110064. [PMID: 31838230 DOI: 10.1016/j.ecoenv.2019.110064] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 11/17/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
Astilbin (ASB), a dihydroflavonol glycoside, is widely found in a variety of plants and in functional foods and acts as a powerful antioxidant. The aim of this study was to investigate the underlying mechanisms involved in the antagonistic effects of ASB on cadmium (Cd)-induced necroptosis in chicken peripheral blood lymphocytes. Peripheral blood lymphocytes were aseptically collected from Roman white hens and then randomly divided into five groups: the control group was incubated without additional reagents, while the other groups were incubated with Cd, ASB, a combination of Cd and ASB, and 0.1% DMSO. After a 24 h treatment, cell samples were collected. The results showed that some morphological changes consistent with necroptosis were observed in the Cd-treated groups, suggesting the occurrence of necroptosis. Simultaneously, antioxidant activity markers (CAT, SOD, GSH, GSH-px, and T-AOC) decreased and indicators of oxidative stress (MDA, iNOS, NO, H2O2, ·OH and ROS) increased. The production of ROS induced the activation of the PI3K/Akt signaling pathway, as the expression levels of PI3K, Akt and PDK1 were significantly elevated. Additionally, the expression levels of RIPK3, RIPK1, MLKL, TAK1, TAB2 and TAB3 were increased and that of Caspase-8 was decreased, which could cause the necroptosis. However, the most important our results was that ASB supplements remarkably attenuated the Cd-induced effects. We conclude that the Cd treatment promoted an imbalance of the antioxidant status and activated the PI3K/Akt pathway, leading to necroptosis in chicken peripheral blood lymphocytes, and that ASB was able to partially ameliorate the effect of Cd-induced necroptosis.
Collapse
Affiliation(s)
- Chi Xin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Shi Guangliang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zhang Qing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Liu Qingqing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Yin Hang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Zhang Yiming
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Li Shu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
27
|
Hu S, Chang X, Zhu H, Wang D, Chen G. PI3K mediates tumor necrosis factor induced-necroptosis through initiating RIP1-RIP3-MLKL signaling pathway activation. Cytokine 2020; 129:155046. [PMID: 32114297 DOI: 10.1016/j.cyto.2020.155046] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 01/15/2023]
Abstract
Necroptosis is a recently identified programmed cell death, which is initiated by receptor-interacting serine/threonine-protein kinase 1 (RIP1), RIP3 and mixed-lineage kinase domain-like protein (MLKL). It has been reported that necroptosis induced by tumor necrosis factor (TNF) was inhibited by the inhibitor of phosphatidylinositol-3-kinase (PI3K) and its substrate protein AKT, indicating that PI3K-AKT signaling pathway was involved in mediating TNF-induced necroptosis, whereas it is unclear how PI3K initiates necroptosis. In this study, we found that TNF-induced necroptosis was inhibited by chemical inhibition or genetic deletion of PI3K. Moreover, knockdown of p110α, the catalytic subunit of PI3K, significantly suppressed the phosphorylation of PI3K substrate protein AKT, and TNF-induced necroptosis was blocked by AKT inhibitors. Furthermore, we found that p110α knockdown also suppressed the phosphorylation and oligomerization of RIP1, RIP3 and MLKL in response to TNF stimulation. In addition to the critical role in mediating TNF-induced necrosome formation, p110α was also essential for the spontaneous phosphorylation of RIP1 and RIP3. Finally, we found that p110α bound to RIP3, but not RIP1, to form protein complex in the process of TNF-induced necroptosis, and mediated TNF-induced necroptosis in the absence of RIP1. Our results demonstrate that PI3K is essential for TNF-induced necroptosis, which may act as the partner of RIP3 to initiate the activation of RIP1-RIP3-MLKL signal pathway and the subsequent necroptosis.
Collapse
Affiliation(s)
- Shiping Hu
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China
| | - Xixi Chang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Hongbin Zhu
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China
| | - Dongxu Wang
- Department of Gastroenterology, The 983rd Hospital of Chinese PLA Joint Logistics Support Force, Tianjin 300142, China.
| | - Guozhu Chen
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| |
Collapse
|
28
|
Muendlein HI, Sarhan J, Liu BC, Connolly WM, Schworer SA, Smirnova I, Tang AY, Ilyukha V, Pietruska J, Tahmasebi S, Sonenberg N, Degterev A, Poltorak A. Constitutive Interferon Attenuates RIPK1/3-Mediated Cytokine Translation. Cell Rep 2020; 30:699-713.e4. [PMID: 31968247 PMCID: PMC7183097 DOI: 10.1016/j.celrep.2019.12.073] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 11/24/2019] [Accepted: 12/18/2019] [Indexed: 01/15/2023] Open
Abstract
Receptor-interacting protein kinase 1 (RIPK1) and 3 (RIPK3) are well known for their capacity to drive necroptosis via mixed-lineage kinase-like domain (MLKL). Recently, RIPK1/3 kinase activity has been shown to drive inflammation via activation of MAPK signaling. However, the regulatory mechanisms underlying this kinase-dependent cytokine production remain poorly understood. In the present study, we establish that the kinase activity of RIPK1/3 regulates cytokine translation in mouse and human macrophages. Furthermore, we show that this inflammatory response is downregulated by type I interferon (IFN) signaling, independent of type I IFN-promoted cell death. Specifically, low-level constitutive IFN signaling attenuates RIPK-driven activation of cap-dependent translation initiation pathway components AKT, mTORC1, 4E-BP and eIF4E, while promoting RIPK-dependent cell death. Altogether, these data characterize constitutive IFN signaling as a regulator of RIPK-dependent inflammation and establish cap-dependent translation as a crucial checkpoint in the regulation of cytokine production.
Collapse
Affiliation(s)
- Hayley I Muendlein
- Graduate Program in Genetics, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Joseph Sarhan
- Medical Scientist Training Program (MSTP), Tufts University School of Medicine, Boston, MA 02111, USA; Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA 02111, USA
| | - Beiyun C Liu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38104, USA
| | - Wilson M Connolly
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Stephen A Schworer
- Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Irina Smirnova
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Amy Y Tang
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Vladimir Ilyukha
- Petrozavodsk State University, Petrozavodsk, Republic of Karelia 185910, Russia
| | - Jodie Pietruska
- Department of Cell, Molecular & Developmental Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Soroush Tahmasebi
- Department of Biochemistry, Goodman Cancer Research Center McGill University, Montreal, QC H3A 1A3, Canada
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Research Center McGill University, Montreal, QC H3A 1A3, Canada
| | - Alexei Degterev
- Department of Cell, Molecular & Developmental Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Alexander Poltorak
- Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA; Petrozavodsk State University, Petrozavodsk, Republic of Karelia 185910, Russia.
| |
Collapse
|
29
|
Wang Z, Feng J, Yu J, Chen G. FKBP12 mediates necroptosis by initiating RIPK1-RIPK3-MLKL signal transduction in response to TNF receptor 1 ligation. J Cell Sci 2019; 132:jcs.227777. [PMID: 31028177 DOI: 10.1242/jcs.227777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
Necroptosis is a regulated form of necrotic cell death that is mediated by receptor-interacting serine/threonine-protein kinase 1 (RIPK1), RIPK3 and mixed-lineage kinase domain-like protein (MLKL), which mediates necroptotic signal transduction induced by tumor necrosis factor (TNF). Although many target proteins for necroptosis have been identified, no report had indicated that FK506-binding protein 12 (FKBP12, also known as FKBP1A), an endogenous protein that regulates protein folding and conformation alteration, is involved in mediating necroptosis. In this study, we found that FKBP12 acts as a novel target protein in mediating necroptosis and the related systemic inflammatory response syndrome triggered by TNF. The mechanistic study discovered that FKBP12 is essential for initiating necrosome formation and RIPK1-RIPK3-MLKL signaling pathway activation in response to TNF receptor 1 ligation. In addition, FKBP12 is indispensable for RIPK1 and RIPK3 expression and subsequent spontaneous phosphorylation, which are essential processes for initial necrosome formation and necroptotic signal transduction; therefore, FKBP12 may target RIPK1 and RIPK3 to mediate necroptosis in vitro and in vivo Collectively, our data demonstrate that FKBP12 could be a potential therapeutic target for the clinical treatment of necroptosis-associated diseases.
Collapse
Affiliation(s)
- Zicheng Wang
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,Beijing Key Laboratory of Therapeutic Gene Engineering Antibody, Beijing 100850, China
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.,Beijing Key Laboratory of Therapeutic Gene Engineering Antibody, Beijing 100850, China
| | - Jiyun Yu
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Guozhu Chen
- Institute of Military Cognition and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| |
Collapse
|
30
|
Abstract
RIPK1 kinase has emerged as a promising therapeutic target for the treatment of a wide range of human neurodegenerative, autoimmune, and inflammatory diseases. This was supported by extensive studies which demonstrated that RIPK1 is a key mediator of apoptotic and necrotic cell death as well as inflammatory pathways. Furthermore, human genetic evidence has linked the dysregulation of RIPK1 to the pathogenesis of ALS as well as other inflammatory and neurodegenerative diseases. Importantly, unique allosteric small-molecule inhibitors of RIPK1 that offer high selectivity have been developed. These molecules can penetrate the blood-brain barrier, thus offering the possibility to target neuroinflammation and cell death which drive various neurologic conditions including Alzheimer's disease, ALS, and multiple sclerosis as well as acute neurological diseases such as stroke and traumatic brain injuries. We discuss the current understanding of RIPK1 regulatory mechanisms and emerging evidence for the pathological roles of RIPK1 in human diseases, especially in the context of the central nervous systems.
Collapse
Affiliation(s)
- Alexei Degterev
- Department of Developmental, Molecular and Chemical Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02445
| | - Dimitry Ofengeim
- Rare and Neurologic Disease Research Therapeutic Area, Sanofi US, Framingham, MA 01701
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
31
|
Park JH, Jung KH, Kim SJ, Yoon YC, Yan HH, Fang Z, Lee JE, Lim JH, Mah S, Hong S, Kim YS, Hong SS. HS-173 as a novel inducer of RIP3-dependent necroptosis in lung cancer. Cancer Lett 2019; 444:94-104. [DOI: 10.1016/j.canlet.2018.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/22/2022]
|
32
|
Escobar SJDM, Fong GM, Winnischofer SMB, Simone M, Munoz L, Dennis JM, Rocha MEM, Witting PK. Anti-proliferative and cytotoxic activities of the flavonoid isoliquiritigenin in the human neuroblastoma cell line SH-SY5Y. Chem Biol Interact 2018; 299:77-87. [PMID: 30502331 DOI: 10.1016/j.cbi.2018.11.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 11/26/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022]
Abstract
Neuroblastoma is a common childhood cancer with high mortality. We evaluated the capacity of the flavonoid, isoliquiritigenin (4,2',4'-trihydroxychalcone; ISL) to inhibit cellular proliferation and migration in the human neuroblastoma cell line SH-SY5Y. Incubation of cultured SH-SY5Y cells with 20-100 μM ISL decreased cell confluency (15-70%) after 24 h incubation, while 10-100 μM ISL (24 h) depleted intracellular ATP stores (15-90% vs vehicle-treated control) after 24 h incubation. ISL-mediated cell toxicity did not involve intracellular caspase 3/7 activation, externalization of phosphatidylserine on the cell membrane or stimulation of TNF and IL-1β release, all indicating that the flavonoid did not induce apoptosis. Pre-treatment of cells with necrostatin-1, a necroptosis inhibitor, significantly restored ATP levels (ATP levels increased 12-42%) in ISL-treated neuroblastoma cells indicative of enhanced viability. By contrast, RIP1 phosphorylation status remained unchanged in cells treated with ISL although the intracellular ratio of phosphorylated/total parental RIP1 increased after ISL treatment on SH-SY5Y cells indicating that ISL decreased levels of native RIP1. In addition, ISL treatment inhibited SH-SY5Y cell migration/proliferation in a scratch assay and arrested cell cycle transition by significantly decreasing the number of cells in G0/G1 phase and increasing populations by ~10% in S (primarily) and G2/M (lesser extent) phases. The intracellular ratio of phosphorylated/total ERK 1/2 and p38 remained unchanged after ISL treatment (up to 40 μM); ERK activation was only determined at ISL dose well above the experimental IC50 value as judged by ELISA analyses and this did not correlate with ISL cytotoxicity at lower dose <40 μM; Western blot assay confirmed the detection of phosphorylated (p-)ERK1/2 and (p-)p38 in ISL treated cells. Together the results suggest that ISL exerts anti-proliferative and cytotoxic activity on SHSY5Y cells through the loss of ATP, induction of cell cycle arrest, and cell death largely via a necroptotic mechanism in the absence of apoptotic activity.
Collapse
Affiliation(s)
- Stephane J de M Escobar
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil; Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Genevieve M Fong
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sheila M B Winnischofer
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Martin Simone
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Lenka Munoz
- Neuropharmacology Groups, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Joanne M Dennis
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Maria Eliane M Rocha
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Paul K Witting
- Redox Biology, Discipline of Pathology, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
33
|
Li X, Huang X, Tang Y, Zhao F, Cao Y, Yin L, Li G. Assessing the Pharmacological and Therapeutic Efficacy of Traditional Chinese Medicine Liangxue Tongyu Prescription for Intracerebral Hemorrhagic Stroke in Neurological Disease Models. Front Pharmacol 2018; 9:1169. [PMID: 30459599 PMCID: PMC6232344 DOI: 10.3389/fphar.2018.01169] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 09/26/2018] [Indexed: 01/17/2023] Open
Abstract
Intracerebral hemorrhage is a fatal subtype of stroke, with crucial impact on public health. Surgical removal of the hematoma as an early-stage treatment for ICH can't improve long-term prognosis remarkably. Liangxue tongyu prescription (LP), a Traditional Chinese Medicine (TCM) formula, includes eight ingredients and has been used to treat ICH in the clinical. In the study, we elucidated the pharmacological efficacy and therapeutic efficacy of LP to dissect the mechanism of LP against ICH via network analysis and experimental validation. First, we discovered 34 potential compounds and 146 corresponding targets in LP based on network prediction. 24 signal pathway were obtained by the Clue Go assay based on potential compounds in LP against ICH. Second, we found that LP can not only decreased the level of high sensitive C reactive protein (HS-CRP), tumor necrosis factor-α (TNF-α), NF-kβ, D-dimmer (D2D), estradiol (E2), S-100B, neuron specific enolase (NSE), and interleukin 1 (IL-1) in plasma on spontaneously hypertensive rats (SHRs), but also promoted cell proliferation and inhibited cell apoptosis on the glutamate-induced PC12 cell. The compounds including Taurine, Paeonol, and Ginsenoside Rb1 in LP can activate PI3K/AKT pathway. Third, from the three-factor two-level factorial design, compound combinations in LP, such as Taurine and Paeonol, Taurine and Geniposide, Ginsenoside Rg1, and Ginsenoside Rb1, had first-level interactions on cell proliferation. Compound combinations including Taurine and Paeonol, Ginsenoside Rg1 and Ginsenoside Rb1 had as significant increase in efficiency on inhibiting the apoptosis of PC12 cells at the low concentration and up-regulating of PI3K and AKT. Overall, our results suggested that LP had integrated therapeutic effect on ICH due to activities of anti-inflammatory, anti-coagulation, blood vessel protection, and protection neuron from excitotoxicity based on the way of "multi-component, multi-target, multi-pathway," and compound combination in LP can offer protection neuron from excitotoxicity at the low concentration by activation of the PI3K/Akt signal pathway.
Collapse
Affiliation(s)
- Xun Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xi Huang
- Jiangsu Wujin Vocational School, Changzhou, China
| | - Yuanlin Tang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fangli Zhao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanmei Cao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lian Yin
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guochun Li
- College of Preclinical Medical, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
34
|
Phenotypic screening identifies a new oxazolone inhibitor of necroptosis and neuroinflammation. Cell Death Discov 2018; 4:10. [PMID: 30062059 PMCID: PMC6060125 DOI: 10.1038/s41420-018-0067-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/06/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022] Open
Abstract
Necroptosis is a regulated form of necrosis, which may be critical in the pathogenesis of neurodegenerative diseases. Neuroinflammation, characterized by the activation of glial cells such as microglia, is closely linked with neurodegenerative pathways and constitutes a major mechanism of neural damage and disease progression. Importantly, inhibition of necroptosis results in disease improvement, unveiling an alternative approach for therapeutic intervention. In the present study, we screened a small library of new molecules, potentially inhibitors of necroptosis, using two cellular models of necroptosis. A new oxazolone, Oxa12, reduced tumour necrosis factor α (TNF-α)-induced necroptosis in mouse L929 fibrosarcoma cells. Notably, Oxa12 strongly inhibited zVAD-fmk-induced necroptosis in murine BV2 microglial cells. Moreover, Oxa12 blocked phosphorylation of mixed-lineage kinase domain-like protein (MLKL), and interfered with necrosome complex formation, indicating that Oxa12 targets components upstream of MLKL. In fact, in silico molecular docking studies revealed that Oxa12 is occupying a region similar to the 1-aminoisoquinoline type II kinase inhibitor inside the receptor-interacting protein 1 (RIP1) kinase domain. Finally, in microglial cells, Oxa12 attenuated zVAD-fmk- and lipopolysaccharide (LPS)-induced inflammatory processes, as revealed by a marked decrease of TNF-α and/or IL-1β expression. More specifically, Oxa12 negatively targeted c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) pathways, as well as NF-κB activation. Overall, we identified a strong lead inhibitor of necroptosis that is also effective at reducing inflammation-associated events. Oxa12 is a promising candidate molecule for further development to target disease states dependent on RIP kinase activity.
Collapse
|
35
|
Jackson TC, Dixon CE, Janesko-Feldman K, Vagni V, Kotermanski SE, Jackson EK, Kochanek PM. Acute Physiology and Neurologic Outcomes after Brain Injury in SCOP/PHLPP1 KO Mice. Sci Rep 2018; 8:7158. [PMID: 29739983 PMCID: PMC5940799 DOI: 10.1038/s41598-018-25371-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/20/2018] [Indexed: 11/12/2022] Open
Abstract
Suprachiasmatic nucleus circadian oscillatory protein (SCOP) (a.k.a. PHLPP1) regulates long-term memory consolidation in the brain. Using a mouse model of controlled cortical impact (CCI) we tested if (1) brain tissue levels of SCOP/PHLPP1 increase after a traumatic brain injury (TBI), and (2) if SCOP/PHLPP1 gene knockout (KO) mice have improved (or worse) neurologic outcomes. Blood chemistry (pH, pCO2, pO2, pSO2, base excess, sodium bicarbonate, and osmolarity) and arterial pressure (MAP) differed in isoflurane anesthetized WT vs. KOs at baseline and up to 1 h post-injury. CCI injury increased cortical/hippocampal SCOP/PHLPP1 levels in WTs 7d and 14d post-injury. Injured KOs had higher brain tissue levels of phosphorylated AKT (pAKT) in cortex (14d post-injury), and higher levels of phosphorylated MEK (pMEK) in hippocampus (7d and 14d post-injury) and in cortex (7d post-injury). Consistent with an important role of SCOP/PHLPP1 on memory function, injured-KOs had near normal performance on the probe trial of the Morris water maze, whereas injured-WTs were impaired. CA1/CA3 hippocampal survival was lower in KOs vs. WTs 24 h post-injury but equivalent by 7d. No difference in 21d cortical lesion volume was detected. SCOP/PHLPP1 overexpression in cultured rat cortical neurons had no effect on 24 h cell death after a mechanical stretch-injury.
Collapse
Affiliation(s)
- Travis C Jackson
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA. .,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA.
| | - C Edward Dixon
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Neurology, 811 Kaufmann Medical Building, 3471 Fifth Avenue, Pittsburgh, USA
| | - Keri Janesko-Feldman
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| | - Vincent Vagni
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| | - Shawn E Kotermanski
- University of Pittsburgh School of Medicine, Department of Pharmacology and Chemical Biology, Bridgeside Point Building 1, 100 Technology Drive, Pittsburgh, USA
| | - Edwin K Jackson
- University of Pittsburgh School of Medicine, Department of Pharmacology and Chemical Biology, Bridgeside Point Building 1, 100 Technology Drive, Pittsburgh, USA
| | - Patrick M Kochanek
- University of Pittsburgh School of Medicine, Safar Center for Resuscitation Research, Children's Hospital of Pittsburgh of UPMC, John G. Rangos Research Center - 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.,University of Pittsburgh School of Medicine, Department of Critical Care Medicine, Scaife Hall, 3550 Terrace Street, Pittsburgh, USA
| |
Collapse
|
36
|
Liu S, Li Y, Choi HMC, Sarkar C, Koh EY, Wu J, Lipinski MM. Lysosomal damage after spinal cord injury causes accumulation of RIPK1 and RIPK3 proteins and potentiation of necroptosis. Cell Death Dis 2018; 9:476. [PMID: 29686269 PMCID: PMC5913300 DOI: 10.1038/s41419-018-0469-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 02/27/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
Necroptosis, a regulated necrosis pathway mediated by the receptor-interacting protein kinases 1 and 3 (RIPK1 and RIPK3), is induced following spinal cord injury (SCI) and thought to contribute to neuronal and glial cell death. However, mechanisms leading to activation of necroptosis after SCI remain unclear. We have previously shown that autophagy, a catabolic pathway facilitating degradation of cytoplasmic proteins and organelles in a lysosome-dependent manner, is inhibited following SCI in rats. Our current data confirm that inhibition of autophagy also occurs after thoracic contusive SCI in the mouse model, as indicated by accumulation of both the autophagosome marker, LC3-II and autophagy cargo protein, p62/SQSTM1. This was most pronounced in the ventral horn neurons and was caused by rapid inhibition of lysosomal function after SCI. Interestingly, RIPK1, RIPK3, and the necroptosis effector protein MLKL also rapidly accumulated after SCI and localized to neurons with disrupted autophagy, suggesting that these events may be related. To determine if lysosomal dysfunction could contribute to induction of necroptosis, we treated PC12 cells and primary rat cortical neurons with lysosomal inhibitors. This led to rapid accumulation of RIPK1 and RIPK3, confirming that they are normally degraded by the lysosomal pathway. In PC12 cells lysosomal inhibition also sensitized cells to necroptosis induced by tumor necrosis factor α (TNFα) and caspase inhibitor. Imaging studies confirmed that RIPK1 partially localized to lysosomes in both untreated and lysosomal inhibitor treated cells. Similarly, we detected presence of RIPK1, RIPK3 and MLKL in both cytosol and at lysosomes after SCI in vivo. Furthermore, stimulation of autophagy and lysosomal function with rapamycin treatment led to decreased accumulation of RIPK1 and attenuated cell death after SCI. These data suggest that lysosomal dysfunction after SCI may contribute to both inhibition of autophagy and sensitize cells to necroptosis by promoting RIPK1 and RIPK3 accumulation.
Collapse
Affiliation(s)
- Shuo Liu
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yun Li
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Harry M C Choi
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chinmoy Sarkar
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eugene Y Koh
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Junfang Wu
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Marta M Lipinski
- Department of Anesthesiology and the Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
37
|
Inhibition of insulin-like growth factor receptor-1 reduces necroptosis-related markers and attenuates LPS-induced lung injury in mice. Biochem Biophys Res Commun 2018; 498:877-883. [PMID: 29545181 DOI: 10.1016/j.bbrc.2018.03.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/09/2018] [Indexed: 02/02/2023]
Abstract
Insulin-like growth factor-1 (IGF-1) levels are known to increase in the bronchoalveolar lavage fluid (BALF) of patients with acute respiratory distress syndrome. Herein, we investigated the role of IGF-1 in lipopolysaccharide (LPS)-induced lung injury. In LPS-treated cells, expressions of receptor-interacting protein 3 (RIP3) and phosphorylated mixed lineage kinase domain-like protein (MLKL) were decreased in IGF-1 receptor small interfering RNA (siRNA)-treated cells compared to control cells. The levels of pro-inflammatory cytokines including interleukin (IL)-1β, IL-6, IL-10, tumour necrosis factor-α, and macrophage inflammatory protein 2/C-X-C motif chemokine ligand 2 in the supernatant were significantly reduced in IGF-1 receptor siRNA-treated cells compared to control cells. In LPS-induced murine lung injury model, total cell counts, polymorphonuclear leukocytes counts, and pro-inflammatory cytokine levels in the BALF were significantly lower and histologically detected lung injury was less common in the group treated with IGF-1 receptor monoclonal antibody compared to the non-treated group. On western blotting, RIP3 and phosphorylated MLKL expressions were relatively decreased in the IGF-1 receptor monoclonal antibody group compared to the non-treated group. IGF-1 may be associated with RIP3-mediated necroptosis in vitro, while blocking of the IGF-1 pathway may reduce LPS-induced lung injuries in vivo.
Collapse
|
38
|
Ogura K, Terasaki Y, Miyoshi-Akiyama T, Terasaki M, Moss J, Noda M, Yahiro K. Vibrio cholerae Cholix Toxin-Induced HepG2 Cell Death is Enhanced by Tumor Necrosis Factor-Alpha Through ROS and Intracellular Signal-Regulated Kinases. Toxicol Sci 2018; 156:455-468. [PMID: 28087840 DOI: 10.1093/toxsci/kfx009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cholix toxin (Cholix) from Vibrio cholerae is a potent virulence factor exhibiting ADP-ribosyltransferase activity on eukaryotic elongation factor 2 (eEF2) of host cells, resulting in the inhibition of protein synthesis. Administration of Cholix or its homologue Pseudomonas exotoxin A (PEA) to mice causes lethal hepatocyte damage. In this study, we demonstrate cytotoxicity of Cholix on human hepatocytes in the presence of tumor necrosis factor α (TNF-α), which has been reported to play a fatal role in PEA administered to mice. Compared with incubating HepG2 cells with Cholix alone, co-treatment with TNF-α and Cholix (TNF-α/Cholix) significantly enhanced the activation of caspases, cytochrome c release from mitochondria into cytoplasm, and poly-ADP-ribose polymerase (PARP) cleavage, while incubation with TNF-α alone or co-treatment with TNF-α/catalytically inactive Cholix did not. In the early stage of cell death, Cholix increased phosphorylation of mitogen-activated protein kinases (e.g., p38, ERK, JNK) and Akt, which was not affected by TNF-α alone. MAPK inhibitors (SP600125, SB20852, and U0126) suppressed PARP cleavage induced by TNF-α/Cholix. Protein kinase inhibitor Go6976 suppressed JNK phosphorylation and PARP cleavage by TNF-α/Cholix. In contrast, PKC activator PMA in the absence of TNF-α promoted Cholix-induced PARP cleavage. Reactive oxygen species (ROS) inhibitor, N-acetyl cysteine (NAC), suppressed TNF-α/Cholix-induced JNK and ERK phosphorylation, resulting in inhibition of PARP cleavage. These data suggest that ROS and JNK pathways are important mediators of TNF-α/Cholix-induced HepG2 cell death.
Collapse
Affiliation(s)
- Kohei Ogura
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuhiro Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Joel Moss
- Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892-1590
| | - Masatoshi Noda
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
39
|
Tiwari R, Singh RD, Khan H, Gangopadhyay S, Mittal S, Singh V, Arjaria N, Shankar J, Roy SK, Singh D, Srivastava V. Oral subchronic exposure to silver nanoparticles causes renal damage through apoptotic impairment and necrotic cell death. Nanotoxicology 2017; 11:671-686. [PMID: 28617070 DOI: 10.1080/17435390.2017.1343874] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Silver nanoparticles (AgNPs) are one of the most widely used nanomaterials. Following oral exposure, AgNPs can accumulate in various organs including kidneys where they show gender specific accumulation. There is limited information on their effect on renal system following long-term animal exposure especially at the ultramicroscopic and molecular level. In this study, we have assessed the effect of 60 days oral AgNPs treatment on kidneys of female Wistar rats at doses of 50 ppm and 200 ppm that are below previously reported lowest observed adverse effect level (LOAEL). AgNPs treatment led to decrease in kidney weight and some loss of renal function as seen by increased levels of serum creatinine and early toxicity markers such as KIM-1, clusterin and osteopontin. We also observed significant mitochondrial damage, loss of brush border membranes, pronounced swelling of podocytes and degeneration of their foot processes using transmission electron microscopy (TEM). These symptoms are similar to those seen in nephrotic syndrome and 'Minimal change disease' of kidney where few changes are visible under light microscopy but significant ultrastructural damage is observed. Prolonged treatment of AgNPs also led to the activation of cell proliferative, survival and proinflammatory factors (Akt/mTOR, JNK/Stat and Erk/NF-κB pathways and IL1β, MIP2, IFN-γ, TNF-α and RANTES) and dysfunction of normal apoptotic pathway. Our study shows how long term AgNPs exposure may promote ultrastructural damage to kidney causing inflammation and expression of cell survival factors. These changes, in the long term, could lead to inhibition of the beneficial apoptotic pathway and promotion of necrotic cell death in kidneys.
Collapse
Affiliation(s)
- Ratnakar Tiwari
- a Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India.,g Department of Biochemistry, School of Dental Sciences , Babu Banarasi Das University Lucknow , Uttar Pradesh , India
| | - Radha Dutt Singh
- a Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Hafizurrahman Khan
- a Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Siddhartha Gangopadhyay
- a Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Sandeep Mittal
- b Nanomaterials Toxicology Laboratory, Nanotherapeutics and Nanomaterial Toxicology Group,CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Vikas Singh
- c Immunotoxicology Laboratory, Food Drug and Chemical Toxicology group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Nidhi Arjaria
- d Advanced Imaging Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Jai Shankar
- d Advanced Imaging Facility, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Somendu Kumar Roy
- e Analytical Chemistry Laboratory , Regulatory Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| | - Dhirendra Singh
- f Animal House Facility, Regulatory Toxicology Group , CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow , Uttar Pradesh , India
| | - Vikas Srivastava
- a Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhavan , Lucknow, India
| |
Collapse
|
40
|
Grootjans S, Vanden Berghe T, Vandenabeele P. Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ 2017; 24:1184-1195. [PMID: 28498367 PMCID: PMC5520172 DOI: 10.1038/cdd.2017.65] [Citation(s) in RCA: 411] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/23/2017] [Accepted: 03/30/2017] [Indexed: 12/17/2022] Open
Abstract
Necroptosis is a form of regulated cell death, which is induced by ligand binding to TNF family death domain receptors, pattern recognizing receptors and virus sensors. The common feature of these receptor systems is the implication of proteins, which contain a receptor interaction protein kinase (RIPK) homology interaction motif (RHIM) mediating recruitment and activation of receptor-interacting protein kinase 3 (RIPK3), which ultimately activates the necroptosis executioner mixed lineage kinase domain-like (MLKL). In case of the TNF family members, the initiator is the survival- and cell death-regulating RIPK1 kinase, in the case of Toll-like receptor 3/4 (TLR3/4), a RHIM-containing adaptor, called TRIF, while in the case of Z-DNA-binding protein ZBP1/DAI, the cytosolic viral sensor itself contains a RHIM domain. In this review, we discuss the different protein complexes that serve as nucleation platforms for necroptosis and the mechanism of execution of necroptosis. Transgenic models (knockout, kinase-dead knock-in) and pharmacologic inhibition indicate that RIPK1, RIPK3 or MLKL are implicated in many inflammatory, degenerative and infectious diseases. However, the conclusion of necroptosis being solely involved in the etiology of diseases is blurred by the pleiotropic roles of RIPK1 and RIPK3 in other cellular processes such as apoptosis and inflammasome activation.
Collapse
Affiliation(s)
- Sasker Grootjans
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Tom Vanden Berghe
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death unit, VIB Inflammation Research Center, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
41
|
Bieri R, Scherr N, Ruf MT, Dangy JP, Gersbach P, Gehringer M, Altmann KH, Pluschke G. The Macrolide Toxin Mycolactone Promotes Bim-Dependent Apoptosis in Buruli Ulcer through Inhibition of mTOR. ACS Chem Biol 2017; 12:1297-1307. [PMID: 28294596 DOI: 10.1021/acschembio.7b00053] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Mycolactone, the macrolide exotoxin produced by Mycobacterium ulcerans, is central to the pathogenesis of the chronic necrotizing skin disease Buruli ulcer (BU). Here we show that mycolactone acts as an inhibitor of the mechanistic Target of Rapamycin (mTOR) signaling pathway by interfering with the assembly of the two distinct mTOR protein complexes mTORC1 and mTORC2, which regulate different cellular processes. Inhibition of the assembly of the rictor containing mTORC2 complex by mycolactone prevents phosphorylation of the serine/threonine protein kinase Akt. The associated inactivation of Akt leads to the dephosphorylation and activation of the Akt-targeted transcription factor FoxO3. Subsequent up-regulation of the FoxO3 target gene BCL2L11 (Bim) increases expression of the pro-apoptotic regulator Bim, driving mycolactone treated mammalian cells into apoptosis. The central role of Bim-dependent apoptosis in BU pathogenesis deduced from our experiments with cultured mammalian cells was further verified in an experimental M. ulcerans infection model. As predicted by the model, M. ulcerans infected Bim knockout mice did not develop necrotic BU lesions with large clusters of extracellular bacteria, but were able to contain the mycobacterial multiplication. Our findings provide a new coherent and comprehensive concept of BU pathogenesis.
Collapse
Affiliation(s)
- Raphael Bieri
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz
1, 4003 Basel, Switzerland
| | - Nicole Scherr
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz
1, 4003 Basel, Switzerland
| | - Marie-Thérèse Ruf
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz
1, 4003 Basel, Switzerland
| | - Jean-Pierre Dangy
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz
1, 4003 Basel, Switzerland
| | - Philipp Gersbach
- Department
of Chemistry and Applied Biosciences, Institute of Pharmaceutical
Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Matthias Gehringer
- Department
of Chemistry and Applied Biosciences, Institute of Pharmaceutical
Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Karl-Heinz Altmann
- Department
of Chemistry and Applied Biosciences, Institute of Pharmaceutical
Sciences, Swiss Federal Institute of Technology (ETH) Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland
| | - Gerd Pluschke
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz
1, 4003 Basel, Switzerland
| |
Collapse
|
42
|
Zhao JX, Yuan YW, Cai CF, Shen DY, Chen ML, Ye F, Mi YJ, Luo QC, Cai WY, Zhang W, Long Y, Zeng Y, Ye GD, Yang SY. Aldose reductase interacts with AKT1 to augment hepatic AKT/mTOR signaling and promote hepatocarcinogenesis. Oncotarget 2017; 8:66987-67000. [PMID: 28978011 PMCID: PMC5620151 DOI: 10.18632/oncotarget.17791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/25/2017] [Indexed: 02/07/2023] Open
Abstract
Marked up-regulation of aldose reductase (AR) is reportedly associated with the development of hepatocellular carcinoma (HCC). We investigated how aberrantly overexpressed AR might promote oncogenic transformation in liver cells and tissues. We found that overexpressed AR interacted with the kinase domain of AKT1 to increase AKT/mTOR signaling. In both cultured liver cancer cells and liver tissues in DEN-induced transgenic HCC model mice, we observed that AR overexpression-induced AKT/mTOR signaling tended to enhance lactate formation and hepatic inflammation to enhance hepatocarcinogenesis. Conversely, AR knockdown suppressed lactate formation and inflammation. Using cultured liver cancer cells, we also demonstrated that AKT1 was essential for AR-induced dysregulation of AKT/mTOR signaling, metabolic reprogramming, antioxidant defense, and inflammatory responses. These findings suggest that aberrantly overexpressed/over-activated hepatic AR promotes HCC development at least in part by interacting with oncogenic AKT1 to augment AKT/mTOR signaling. Inhibition of AR and/or AKT1 might serve as an effective strategy for the prevention and therapy of liver cancer.
Collapse
Affiliation(s)
- Jia-Xing Zhao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361002, China
| | - Ya-Wei Yuan
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, Fujian, 361002, China
| | - Cheng-Fu Cai
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Dong-Yan Shen
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Mao-Li Chen
- School of Pharmaceutical Science, Xiamen University, Xiamen, Fujian, 361003, China
| | - Feng Ye
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Yan-Jun Mi
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Qi-Cong Luo
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Wang-Yu Cai
- Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Wei Zhang
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Ying Long
- Translational Medicine Center, Hunan Cancer Hospital, Changsha, Hunan, 410013, China
| | - Yong Zeng
- Translational Medicine Center, Hunan Cancer Hospital, Changsha, Hunan, 410013, China
| | - Guo-Dong Ye
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| | - Shu-Yu Yang
- The First Affiliated Hospital, Medical College, Xiamen University, Xiamen, Fujian, 361003, China
| |
Collapse
|
43
|
Saleh D, Najjar M, Zelic M, Shah S, Nogusa S, Polykratis A, Paczosa MK, Gough PJ, Bertin J, Whalen M, Fitzgerald KA, Slavov N, Pasparakis M, Balachandran S, Kelliher M, Mecsas J, Degterev A. Kinase Activities of RIPK1 and RIPK3 Can Direct IFN-β Synthesis Induced by Lipopolysaccharide. THE JOURNAL OF IMMUNOLOGY 2017; 198:4435-4447. [PMID: 28461567 DOI: 10.4049/jimmunol.1601717] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
Abstract
The innate immune response is a central element of the initial defense against bacterial and viral pathogens. Macrophages are key innate immune cells that upon encountering pathogen-associated molecular patterns respond by producing cytokines, including IFN-β. In this study, we identify a novel role for RIPK1 and RIPK3, a pair of homologous serine/threonine kinases previously implicated in the regulation of necroptosis and pathologic tissue injury, in directing IFN-β production in macrophages. Using genetic and pharmacologic tools, we show that catalytic activity of RIPK1 directs IFN-β synthesis induced by LPS in mice. Additionally, we report that RIPK1 kinase-dependent IFN-β production may be elicited in an analogous fashion using LPS in bone marrow-derived macrophages upon inhibition of caspases. Notably, this regulation requires kinase activities of both RIPK1 and RIPK3, but not the necroptosis effector protein, MLKL. Mechanistically, we provide evidence that necrosome-like RIPK1 and RIPK3 aggregates facilitate canonical TRIF-dependent IFN-β production downstream of the LPS receptor TLR4. Intriguingly, we also show that RIPK1 and RIPK3 kinase-dependent synthesis of IFN-β is markedly induced by avirulent strains of Gram-negative bacteria, Yersinia and Klebsiella, and less so by their wild-type counterparts. Overall, these observations identify unexpected roles for RIPK1 and RIPK3 kinases in the production of IFN-β during the host inflammatory responses to bacterial infection and suggest that the axis in which these kinases operate may represent a target for bacterial virulence factors.
Collapse
Affiliation(s)
- Danish Saleh
- Medical Scientist Training Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111.,Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Malek Najjar
- Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Matija Zelic
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Saumil Shah
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111
| | - Shoko Nogusa
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine, University of Cologne, 50674 Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany
| | - Michelle K Paczosa
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426
| | - Michael Whalen
- Department of Pediatric Critical Care Medicine, Neuroscience Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605
| | - Nikolai Slavov
- Department of Bioengineering and Biology, Northeastern University, Boston, MA 02115; and
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany.,Center for Molecular Medicine, University of Cologne, 50674 Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Michelle Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111
| | - Alexei Degterev
- Medical Scientist Training Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111; .,Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111.,Graduate Program in Pharmacology and Experimental Therapeutics, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111.,Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|
44
|
Wegner KW, Saleh D, Degterev A. Complex Pathologic Roles of RIPK1 and RIPK3: Moving Beyond Necroptosis. Trends Pharmacol Sci 2017; 38:202-225. [PMID: 28126382 DOI: 10.1016/j.tips.2016.12.005] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/09/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023]
Abstract
A process of regulated necrosis, termed necroptosis, has been recognized as a major contributor to cell death and inflammation occurring under a wide range of pathologic settings. The core event in necroptosis is the formation of the detergent-insoluble 'necrosome' complex of homologous Ser/Thr kinases, receptor protein interacting kinase 1 (RIPK1) and receptor interacting protein kinase 3 (RIPK3), which promotes phosphorylation of a key prodeath effector, mixed lineage kinase domain-like (MLKL), by RIPK3. Core necroptosis mediators are under multiple controls, which have been a subject of intense investigation. Additional, non-necroptotic functions of these factors, primarily in controlling apoptosis and inflammatory responses, have also begun to emerge. This review will provide an overview of the current understanding of the human disease relevance of this pathway, and potential therapeutic strategies, targeting necroptosis mediators in various pathologies.
Collapse
Affiliation(s)
- Kelby W Wegner
- Master of Science in Biomedical Sciences Program, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Danish Saleh
- Medical Scientist Training Program and Program in Neuroscience, Sackler Graduate School, Tufts University, Boston, MA 02111, USA
| | - Alexei Degterev
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA.
| |
Collapse
|
45
|
Dual PI3K/ERK inhibition induces necroptotic cell death of Hodgkin Lymphoma cells through IER3 downregulation. Sci Rep 2016; 6:35745. [PMID: 27767172 PMCID: PMC5073341 DOI: 10.1038/srep35745] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 10/04/2016] [Indexed: 12/18/2022] Open
Abstract
PI3K/AKT and RAF/MEK/ERK pathways are constitutively activated in Hodgkin lymphoma (HL) patients, thus representing attractive therapeutic targets. Here we report that the PI3K/ERK dual inhibitor AEZS-136 induced significant cell proliferation inhibition in L-540, SUP-HD1, KM-H2 and L-428 HL cell lines, but a significant increase in necroptotic cell death was observed only in two out of four cell lines (L-540 and SUP-HD1). In these cells, AEZS-136-induced necroptosis was associated with mitochondrial dysfunction and reactive oxygen species (ROS) production. JNK was activated by AEZS-136, and AEZS-136-induced necroptosis was blocked by the necroptosis inhibitor necrostatin-1 or the JNK inhibitor SP600125, suggesting that JNK activation is required to trigger necroptosis following dual PI3K/ERK inhibition. Gene expression analysis indicated that the effects of AEZS-136 were associated with the modulation of cell cycle and cell death pathways. In the cell death-resistant cell lines, AEZS-136 induced the expression of immediate early response 3 (IER3) both in vitro and in vivo. Silencing of IER3 restored sensitivity to AEZS-136-induced necroptosis. Furthermore, xenograft studies demonstrated a 70% inhibition of tumor growth and a 10-fold increase in tumor necrosis in AEZS-136-treated animals. Together, these data suggest that dual PI3K/ERK inhibition might be an effective approach for improving therapeutic outcomes in HL.
Collapse
|
46
|
Najjar M, Saleh D, Zelic M, Nogusa S, Shah S, Tai A, Finger JN, Polykratis A, Gough PJ, Bertin J, Whalen M, Pasparakis M, Balachandran S, Kelliher M, Poltorak A, Degterev A. RIPK1 and RIPK3 Kinases Promote Cell-Death-Independent Inflammation by Toll-like Receptor 4. Immunity 2016; 45:46-59. [PMID: 27396959 DOI: 10.1016/j.immuni.2016.06.007] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 03/14/2016] [Accepted: 04/05/2016] [Indexed: 01/28/2023]
Abstract
Macrophages are a crucial component of the innate immune system in sensing pathogens and promoting local and systemic inflammation. RIPK1 and RIPK3 are homologous kinases, previously linked to activation of necroptotic death. In this study, we have described roles for these kinases as master regulators of pro-inflammatory gene expression induced by lipopolysaccharide, independent of their well-documented cell death functions. In primary macrophages, this regulation was elicited in the absence of caspase-8 activity, required the adaptor molecule TRIF, and proceeded in a cell autonomous manner. RIPK1 and RIPK3 kinases promoted sustained activation of Erk, cFos, and NF-κB, which were required for inflammatory changes. Utilizing genetic and pharmacologic tools, we showed that RIPK1 and RIPK3 account for acute inflammatory responses induced by lipopolysaccharide in vivo; notably, this regulation did not require exogenous manipulation of caspases. These findings identified a new pharmacologically accessible pathway that may be relevant to inflammatory pathologies.
Collapse
Affiliation(s)
- Malek Najjar
- Program in Pharmacology and Experimental Therapeutics, Sackler Graduate School, Tufts University, Boston, MA 02111, USA
| | - Danish Saleh
- Medical Scientist Training Program and Program in Neuroscience, Sackler Graduate School, Tufts University, Boston, MA 02111, USA
| | - Matija Zelic
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shoko Nogusa
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Saumil Shah
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Albert Tai
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Joshua N Finger
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Apostolos Polykratis
- Institute for Genetics, Center for Molecular Medicine and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany
| | - Peter J Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline, Collegeville, PA 19426, USA
| | - Michael Whalen
- Neuroscience Center and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Manolis Pasparakis
- Institute for Genetics, Center for Molecular Medicine and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Michelle Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Alexander Poltorak
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Alexei Degterev
- Program in Pharmacology and Experimental Therapeutics, Sackler Graduate School, Tufts University, Boston, MA 02111, USA.,Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
47
|
Tang X, Amar S. Kavain Involvement in LPS-Induced Signaling Pathways. J Cell Biochem 2016; 117:2272-80. [PMID: 26917453 DOI: 10.1002/jcb.25525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/22/2016] [Indexed: 12/17/2022]
Abstract
Kavain, a compound extracted from the Kava plant, Piper methysticum, is found to be involved in TNF-α expression in human and mouse cells via regulation of transcriptional factors such as NF-kB and LITAF. LITAF is known to activate the transcription of more than 20 cytokines that are involved in a variety of cellular processes and is associated with many inflammatory diseases, including angiogenesis, cancer, arthritis, and more. The modulation of LITAF is expected to positively affect cytokine-mediated diseases. Thus, intensive efforts have been deployed in search of LITAF inhibitors. In this work, we found that, in vitro, Kavain reduced LPS- induced TNF-α secretion in mouse macrophages, mouse bone marrow macrophages (BMM), and human peripheral blood mononuclear cells (HPBMC). We also found that Kavain treatment in RAW264.7 cells deactivated MyD88 and Akt, inhibited LITAF, and reduced the production of TNF-α, IL-27, and MIG in response to LPS. Similarly, it had a significant in vivo anti-inflammatory effect on wild-type (WT) mice that developed Collagen Antibody Induced Arthritis (CAIA). Overall, MyD88 was found to be an important mediator of the LPS-induced inflammatory response that can be distinguished from the NF-κB pathway. We also found that MyD88 is involved in the pathway linking LPS/LITAF to TNF-α. Therefore, given that Kavain modulates LPS-induced signaling pathways leading to cytokine expression, therapeutic interventions involving Kavain in inflammatory diseases are warranted. J. Cell. Biochem. 117: 2272-2280, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaoren Tang
- Department of Molecular and Cell Biology, Center for Anti-Inflammatory Therapeutics, Boston University Goldman School of Dental Medicine, Boston, 02118, Massachusetts
| | - Salomon Amar
- Department of Molecular and Cell Biology, Center for Anti-Inflammatory Therapeutics, Boston University Goldman School of Dental Medicine, Boston, 02118, Massachusetts
| |
Collapse
|
48
|
Inhibition of Receptor Interacting Protein Kinases Attenuates Cardiomyocyte Hypertrophy Induced by Palmitic Acid. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1451676. [PMID: 27057269 PMCID: PMC4736315 DOI: 10.1155/2016/1451676] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 02/05/2023]
Abstract
Palmitic acid (PA) is known to cause cardiomyocyte dysfunction. Cardiac hypertrophy is one of the important pathological features of PA-induced lipotoxicity, but the mechanism by which PA induces cardiomyocyte hypertrophy is still unclear. Therefore, our study was to test whether necroptosis, a receptor interacting protein kinase 1 and 3 (RIPK1 and RIPK3-) dependent programmed necrosis, was involved in the PA-induced cardiomyocyte hypertrophy. We used the PA-treated primary neonatal rat cardiac myocytes (NCMs) or H9c2 cells to study lipotoxicity. Our results demonstrated that cardiomyocyte hypertrophy was induced by PA treatment, determined by upregulation of hypertrophic marker genes and cell surface area enlargement. Upon PA treatment, the expression of RIPK1 and RIPK3 was increased. Pretreatment with the RIPK1 inhibitor necrostatin-1 (Nec-1), the PA-induced cardiomyocyte hypertrophy, was attenuated. Knockdown of RIPK1 or RIPK3 by siRNA suppressed the PA-induced myocardial hypertrophy. Moreover, a crosstalk between necroptosis and endoplasmic reticulum (ER) stress was observed in PA-treated cardiomyocytes. Inhibition of RIPK1 with Nec-1, phosphorylation level of AKT (Ser473), and mTOR (Ser2481) was significantly reduced in PA-treated cardiomyocytes. In conclusion, RIPKs-dependent necroptosis might be crucial in PA-induced myocardial hypertrophy. Activation of mTOR may mediate the effect of necroptosis in cardiomyocyte hypertrophy induced by PA.
Collapse
|
49
|
Jia Q, Cheng W, Yue Y, Hu Y, Zhang J, Pan X, Xu Z, Zhang P. Cucurbitacin E inhibits TNF-α-induced inflammatory cytokine production in human synoviocyte MH7A cells via suppression of PI3K/Akt/NF-κB pathways. Int Immunopharmacol 2015; 29:884-890. [DOI: 10.1016/j.intimp.2015.08.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 01/29/2023]
|
50
|
Structure guided design of potent and selective ponatinib-based hybrid inhibitors for RIPK1. Cell Rep 2015; 10:1850-60. [PMID: 25801024 DOI: 10.1016/j.celrep.2015.02.052] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 01/13/2015] [Accepted: 02/23/2015] [Indexed: 12/27/2022] Open
Abstract
RIPK1 and RIPK3, two closely related RIPK family members, have emerged as important regulators of pathologic cell death and inflammation. In the current work, we report that the Bcr-Abl inhibitor and anti-leukemia agent ponatinib is also a first-in-class dual inhibitor of RIPK1 and RIPK3. Ponatinib potently inhibited multiple paradigms of RIPK1- and RIPK3-dependent cell death and inflammatory tumor necrosis factor alpha (TNF-α) gene transcription. We further describe design strategies that utilize the ponatinib scaffold to develop two classes of inhibitors (CS and PN series), each with greatly improved selectivity for RIPK1. In particular, we detail the development of PN10, a highly potent and selective "hybrid" RIPK1 inhibitor, capturing the best properties of two different allosteric RIPK1 inhibitors, ponatinib and necrostatin-1. Finally, we show that RIPK1 inhibitors from both classes are powerful blockers of TNF-induced injury in vivo. Altogether, these findings outline promising candidate molecules and design approaches for targeting RIPK1- and RIPK3-driven inflammatory pathologies.
Collapse
|