151
|
Zhang X, Kitatani K, Toyoshima M, Ishibashi M, Usui T, Minato J, Egiz M, Shigeta S, Fox T, Deering T, Kester M, Yaegashi N. Ceramide Nanoliposomes as a MLKL-Dependent, Necroptosis-Inducing, Chemotherapeutic Reagent in Ovarian Cancer. Mol Cancer Ther 2018; 17:50-59. [PMID: 29079707 PMCID: PMC5752574 DOI: 10.1158/1535-7163.mct-17-0173] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/28/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
Ceramides are bioactive lipids that mediate cell death in cancer cells, and ceramide-based therapy is now being tested in dose-escalating phase I clinical trials as a cancer treatment. Multiple nanoscale delivery systems for ceramide have been proposed to overcome the inherent toxicities, poor pharmacokinetics, and difficult biophysics associated with ceramide. Using the ceramide nanoliposomes (CNL), we now investigate the therapeutic efficacy and signaling mechanisms of this nanoscale delivery platform in refractory ovarian cancer. Treatment of ovarian cancer cells with CNL decreased the number of living cells through necroptosis but not apoptosis. Mechanistically, dying SKOV3 ovarian cancer cells exhibit activation of pseudokinase mixed lineage kinase domain-like (MLKL) as evidenced by oligomerization and relocalization to the blebbing membranes, showing necroptotic characteristics. Knockdown of MLKL, but not its upstream protein kinases such as receptor-interacting protein kinases, with siRNA significantly abolished CNL-induced cell death. Monomeric MLKL protein expression inversely correlated with the IC50 values of CNL in distinct ovarian cancer cell lines, suggesting MLKL as a possible determinant for CNL-induced cell death. Finally, systemic CNL administration suppressed metastatic growth in an ovarian cancer cell xenograft model. Taken together, these results suggest that MLKL is a novel pronecroptotic target for ceramide in ovarian cancer models. Mol Cancer Ther; 17(1); 50-59. ©2017 AACR.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Kazuyuki Kitatani
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan.
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masafumi Toyoshima
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan.
| | - Masumi Ishibashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Toshinori Usui
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Junko Minato
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Mahy Egiz
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Shogo Shigeta
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
| | - Todd Fox
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Tye Deering
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| |
Collapse
|
152
|
Varfolomeev E, Vucic D. Intracellular regulation of TNF activity in health and disease. Cytokine 2018; 101:26-32. [DOI: 10.1016/j.cyto.2016.08.035] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 01/27/2023]
|
153
|
Zhang S, Qian C, Liu X, Piao S, Jin X. TRAF6 Affects RAC1 Expression and Apoptosis in SK-Hep1 Cells. Chin Med 2018. [DOI: 10.4236/cm.2018.94011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
154
|
Hancz D, Szabo A, Molnar T, Varga Z, Hancz A, Gregus A, Hueber AO, Rajnavolgyi E, Koncz G. Flagellin increases death receptor-mediated cell death in a RIP1-dependent manner. Immunol Lett 2018; 193:42-50. [DOI: 10.1016/j.imlet.2017.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/18/2017] [Accepted: 11/20/2017] [Indexed: 12/26/2022]
|
155
|
Affiliation(s)
- Rimpy Dhingra
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre , Winnipeg, Manitoba , Canada
| | - Amir Ravandi
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre , Winnipeg, Manitoba , Canada
| | - Lorrie A Kirshenbaum
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre , Winnipeg, Manitoba , Canada.,Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba , Winnipeg, Manitoba , Canada
| |
Collapse
|
156
|
Key roles of necroptotic factors in promoting tumor growth. Oncotarget 2017; 7:22219-33. [PMID: 26959742 PMCID: PMC5008357 DOI: 10.18632/oncotarget.7924] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/23/2016] [Indexed: 02/05/2023] Open
Abstract
Necroptotic factors are generally assumed to play a positive role in tumor therapy by eliminating damaged tumor cells. Here we show that, contrary to expectation, necroptotic factors RIPK1, RIPK3, and MLKL promote tumor growth. We demonstrate that genetic knockout of necroptotic genes RIPK1, RIPK3, or MLKL in cancer cells significantly attenuated their abilities to grow in an anchorage-independent manner. In addition, they exhibited significantly enhanced radiosensitivity. The knockout cells also showed greatly reduced ability to form tumors in mice. Moreover, necrosulfonamide (NSA), a previously identified chemical inhibitor of necroptosis, could significantly delay tumor growth in a xenograft model. Mechanistically, we show that necroptoic factors play a significant role in maintaining the activity of NF-κB. Finally, we found that high levels of phosphorylated MLKL in human esophageal and colon cancers are associated with poor overall survival. Taken together, we conclude that pro-necroptic factors such as RIPK1, RIPK3, and MLKL may play a role in supporting tumor growth, and MLKL may be a promising target for cancer treatment.
Collapse
|
157
|
Chang X, Wang L, Wang Z, Wu S, Zhu X, Hu S, Wang Y, Yu J, Chen G. TRADD mediates the tumor necrosis factor-induced apoptosis of L929 cells in the absence of RIP3. Sci Rep 2017; 7:16111. [PMID: 29170425 PMCID: PMC5701027 DOI: 10.1038/s41598-017-16390-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/09/2017] [Indexed: 01/01/2023] Open
Abstract
Receptor-interacting protein kinase 3 (RIP3) is a critical initiator in mediating necroptosis induced by tumor necrosis factor alpha (TNFα) in L929 cells, so knockdown of RIP3 inhibits TNFα-induced L929 cell necroptosis. However, RIP3 knockdown was shown to switch TNFα-induced necroptosis to apoptosis in L929 cells in other studies. Therefore, whether RIP3 knockdown blocks the TNFα-induced death of L929 cells is controversial. In this study, TNFα activated caspase pathway and induced cell death in RIP3 knockdown L929 cells, and the RIP3-independent cell death had been blocked by Z-VAD-FMK (pan-caspase inhibitor) or caspase 8 knockdown, demonstrating that RIP3 knockdown switched TNFα-induced necroptosis to caspase-dependent apoptosis. Although both TNF receptor type 1-associated death domain protein (TRADD) and RIP1 have been reported to mediate TNFα-induced apoptosis, the knockdown of TRADD, but not RIP1, suppressed TNFα-induced activation of the caspase pathway and subsequent apoptosis in RIP3 knockdown L929 cells. In addition, TRADD bound and activated caspase 8 during the RIP3-independent apoptosis process, indicating that TRADD initiates RIP3-independent apoptosis by activating the caspase pathway. Collectively, we identified the target and mechanism underlying RIP3-independent apoptosis and elucidated the coordinated roles of RIP3 and TRADD in mediating the programmed cell death of L929 cells following TNFα stimulation.
Collapse
Affiliation(s)
- Xixi Chang
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Lili Wang
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Zicheng Wang
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Shuai Wu
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Xiaoming Zhu
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Shiping Hu
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China
| | - Yu Wang
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China.
| | - Jiyun Yu
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China.
| | - Guozhu Chen
- Department of Frontier for Biological Treatment, Beijing Institute of Basic Medical Science, Beijing, 100850, China.
| |
Collapse
|
158
|
Zhao X, Khan N, Gan H, Tzelepis F, Nishimura T, Park SY, Divangahi M, Remold HG. Bcl-x L mediates RIPK3-dependent necrosis in M. tuberculosis-infected macrophages. Mucosal Immunol 2017; 10:1553-1568. [PMID: 28401933 PMCID: PMC5638669 DOI: 10.1038/mi.2017.12] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 01/13/2017] [Indexed: 02/04/2023]
Abstract
Virulent Mycobacterium tuberculosis (Mtb) triggers necrosis in host Mϕ, which is essential for successful pathogenesis in tuberculosis. Here we demonstrate that necrosis of Mtb-infected Mϕ is dependent on the action of the cytosolic Receptor Interacting Protein Kinase 3 (RIPK3) and the mitochondrial Bcl-2 family member protein B-cell lymphoma-extra large (Bcl-xL). RIPK3-deficient Mϕ are able to better control bacterial growth in vitro and in vivo. Mechanistically, cytosolic RIPK3 translocates to the mitochondria where it promotes necrosis and blocks caspase 8-activation and apoptosis via Bcl-xL. Furthermore, necrosis is associated with stabilization of hexokinase II on the mitochondria as well as cyclophilin D-dependent mitochondrial permeability transition. Collectively, these events upregulate the level of reactive oxygen species to induce necrosis. Thus, in Mtb-infected Mϕ, mitochondria are an essential platform for induction of necrosis by activating RIPK3 function and preventing caspase 8-activation.
Collapse
Affiliation(s)
- Xiaomin Zhao
- Division of Rheumatology, Immunology and Allergy, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
| | - Nargis Khan
- Department of Medicine, Department of Microbiology & Immunology,
McGill International TB Centre, McGill University Health Centre, Meakins-Christie
Laboratories, Montreal, Quebec, H4A 3J1, Canada
| | - Huixian Gan
- Division of Rheumatology, Immunology and Allergy, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
| | - Fanny Tzelepis
- Department of Medicine, Department of Microbiology & Immunology,
McGill International TB Centre, McGill University Health Centre, Meakins-Christie
Laboratories, Montreal, Quebec, H4A 3J1, Canada
| | - Tomoyasu Nishimura
- Division of Rheumatology, Immunology and Allergy, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
- Health Center, Keio University. 35 Shinamo machi; Tokyo 160-8582,
Japan
| | - Seung-Yeol Park
- Division of Rheumatology, Immunology and Allergy, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
| | - Maziar Divangahi
- Department of Medicine, Department of Microbiology & Immunology,
McGill International TB Centre, McGill University Health Centre, Meakins-Christie
Laboratories, Montreal, Quebec, H4A 3J1, Canada
| | - Heinz G. Remold
- Division of Rheumatology, Immunology and Allergy, Department of
Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA,
USA
| |
Collapse
|
159
|
Jiang JZ, Ye J, Jin GY, Piao HM, Cui H, Zheng MY, Yang JS, Che N, Choi YH, Li LC, Yan GH. Asiaticoside Mitigates the Allergic Inflammation by Abrogating the Degranulation of Mast Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:8128-8135. [PMID: 28891650 DOI: 10.1021/acs.jafc.7b01590] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The effects of asiaticoside (AS) on allergic responses mediated by mast cells were investigated. AS showed no obvious cytotoxicity on RPMCs (rat peritoneal mast cells). AS reduced the intracellular calcium in RPMCs and deprived the histamine release and degranulation. AS also decreased the generation of antigen-induced tumor necrosis factor α, interleukin (IL)-4, IL-8, and IL-1β in RBL-2H3 cells sensitized by IgE. The suppression of AS on pro-inflammatory cytokines was related with the activation of the intracellular FcεRI and the inhibition of the nuclear factor-κB signaling pathway. In addition, AS disabled the phosphorylation of antigen-induced Syk, Lyn, Gab2, and PLCγ1, thus suppressing the downstream Akt phosphorylation and MAPKs pathways. It also increased HO-1 and Nrf2 expression time dependently. In summary, we demonstrate that AS suppresses the allergic inflammation mediated by mast cells and this effect might be mediated by FcεRI-dependent signaling pathways.
Collapse
Affiliation(s)
- Jing Zhi Jiang
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| | - Jing Ye
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| | - Guang Yu Jin
- Department of Respiratory Medicine, Yanbian University Hospital , Yanji 133000, Jilin China
| | - Hong Mei Piao
- Department of Respiratory Medicine, Yanbian University Hospital , Yanji 133000, Jilin China
| | - Hong Cui
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| | - Ming Yu Zheng
- College of Pharmacy, Yanbian University , Yanji 133002, Jilin, China
| | - Jin Shi Yang
- College of Pharmacy, Yanbian University , Yanji 133002, Jilin, China
| | - Nan Che
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| | - Yun Ho Choi
- Department of Anatomy, Medical School of Institute of Medical Sciences, Chonbuk National University , Jeonju, Jeonbuk 561-756, Republic of Korea
| | - Liang Chang Li
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| | - Guang Hai Yan
- Department of Anatomy, Histology and Embryology, Medical College of Yanbian University , Yanji 133002, Jilin, China
| |
Collapse
|
160
|
Ghandadi M, Behravan J, Abnous K, Ehtesham Gharaee M, Mosaffa F. TNF-α exerts cytotoxic effects on multidrug resistant breast cancer MCF-7/MX cells via a non-apoptotic death pathway. Cytokine 2017. [PMID: 28651126 DOI: 10.1016/j.cyto.2017.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
161
|
Zhou K, Shi L, Wang Z, Zhou J, Manaenko A, Reis C, Chen S, Zhang J. RIP1-RIP3-DRP1 pathway regulates NLRP3 inflammasome activation following subarachnoid hemorrhage. Exp Neurol 2017; 295:116-124. [PMID: 28579326 DOI: 10.1016/j.expneurol.2017.06.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/21/2017] [Accepted: 06/01/2017] [Indexed: 12/30/2022]
Abstract
The NLRP3 inflammasome functions as a crucial component of the inflammatory response in early brain injury (EBI) after subarachnoid hemorrhage (SAH). However, the mechanisms underlying the activation of NLRP3 inflammasome has not been well elucidated. In this study, we hypothesized the RIP1-RIP3-DRP1 pathway was involved in the activation of the NLRP3 inflammasome following SAH. SAH was induced by endovascular perforation in rats. Necrostatin-1 (Nec-1) or mitochondrial division inhibitor (Mdivi-1) was administered 1h after SAH by intraperitoneal injection. SAH grade, neurological function, brain water content, Western blot, ROS assay, immunofluorescence and transmission electron microscopy were performed. SAH led to the upregulation of RIP1, RIP3, phosphorylated DRP1 and NLRP3 inflammasome. Nec-1 treatment reduced RIP1, RIP3, phosphorylated DRP1 and NLRP3 inflammasome, subsequently alleviated brain edema and neurological deficits at 24h following SAH. The treatment with Mdivi-1 inhibited the expression of DRP1 protein, attenuated mitochondria damage and the generation of ROS, inhibited NLRP3 inflammasome and ameliorated brain edema and neurological deficits at 24h after SAH. The activation of the NLRP3 inflammasome in EBI after SAH was mediated by RIP1-RIP3-DRP1 pathway. Nec-1 and Mdivi-1 can inhibit inflammation and improve neurological function after SAH.
Collapse
Affiliation(s)
- Keren Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingyi Zhou
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Sheng Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Brain research institute, Zhejiang University, Hangzhou, Zhejiang, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
162
|
Goodall ML, Fitzwalter BE, Zahedi S, Wu M, Rodriguez D, Mulcahy-Levy JM, Green DR, Morgan M, Cramer SD, Thorburn A. The Autophagy Machinery Controls Cell Death Switching between Apoptosis and Necroptosis. Dev Cell 2017; 37:337-349. [PMID: 27219062 DOI: 10.1016/j.devcel.2016.04.018] [Citation(s) in RCA: 250] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 03/29/2016] [Accepted: 04/25/2016] [Indexed: 12/18/2022]
Abstract
Although autophagy controls cell death and survival, underlying mechanisms are poorly understood, and it is unknown whether autophagy affects only whether or not cells die or also controls other aspects of programmed cell death. MAP3K7 is a tumor suppressor gene associated with poor disease-free survival in prostate cancer. Here, we report that Map3k7 deletion in mouse prostate cells sensitizes to cell death by TRAIL (TNF-related apoptosis-inducing ligand). Surprisingly, this death occurs primarily through necroptosis, not apoptosis, due to assembly of the necrosome in association with the autophagy machinery, mediated by p62/SQSTM1 recruitment of RIPK1. The mechanism of cell death switches to apoptosis if p62-dependent recruitment of the necrosome to the autophagy machinery is blocked. These data show that the autophagy machinery can control the mechanism of programmed cell death by serving as a scaffold rather than by degrading cargo.
Collapse
Affiliation(s)
- Megan L Goodall
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Brent E Fitzwalter
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Shadi Zahedi
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Min Wu
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA; Department of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Diego Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jean M Mulcahy-Levy
- Department of Pediatrics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael Morgan
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Scott D Cramer
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
163
|
Ishii T, Kawakami E, Endo K, Misawa H, Watabe K. Formation and spreading of TDP-43 aggregates in cultured neuronal and glial cells demonstrated by time-lapse imaging. PLoS One 2017; 12:e0179375. [PMID: 28599005 PMCID: PMC5466347 DOI: 10.1371/journal.pone.0179375] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 05/30/2017] [Indexed: 02/07/2023] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a main constituent of cytoplasmic aggregates in neuronal and glial cells in cases of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We have previously demonstrated that adenovirus-transduced artificial TDP-43 cytoplasmic aggregates formation is enhanced by proteasome inhibition in vitro and in vivo. However, the relationship between cytoplasmic aggregate formation and cell death remains unclear. In the present study, rat neural stem cell lines stably transfected with EGFP- or Sirius-expression vectors under the control of tubulin beta III, glial fibrillary acidic protein, or 2',3'-cyclic nucleotide 3'-phosphodiesterase promoter were differentiated into neurons, astrocytes, and oligodendrocytes, respectively, in the presence of retinoic acid. The differentiated cells were then transduced with adenoviruses expressing DsRed-tagged human wild type and C-terminal fragment TDP-43 under the condition of proteasome inhibition. Time-lapse imaging analyses revealed growing cytoplasmic aggregates in the transduced neuronal and glial cells, followed by collapse of the cell. The aggregates remained insoluble in culture media, consisted of sarkosyl-insoluble granular materials, and contained phosphorylated TDP-43. Moreover, the released aggregates were incorporated into neighboring neuronal cells, suggesting cell-to-cell spreading. The present study provides a novel tool for analyzing the detailed molecular mechanisms of TDP-43 proteinopathy in vitro.
Collapse
Affiliation(s)
- Tomohiro Ishii
- Laboratory for Neurodegenerative Pathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Emiko Kawakami
- Laboratory for Neurodegenerative Pathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kentaro Endo
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hidemi Misawa
- Division of Pharmacology, Faculty of Pharmacy, Keio University, Tokyo, Japan
- * E-mail: (HM); (KW)
| | - Kazuhiko Watabe
- Laboratory for Neurodegenerative Pathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Medical Technology (Neuropathology), Kyorin University Faculty of Health Sciences, Tokyo, Japan
- * E-mail: (HM); (KW)
| |
Collapse
|
164
|
Choong OK, Lee DS, Chen CY, Hsieh PCH. The roles of non-coding RNAs in cardiac regenerative medicine. Noncoding RNA Res 2017; 2:100-110. [PMID: 30159427 PMCID: PMC6096405 DOI: 10.1016/j.ncrna.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 06/03/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023] Open
Abstract
The emergence of non-coding RNAs (ncRNAs) has challenged the central dogma of molecular biology that dictates that the decryption of genetic information starts from transcription of DNA to RNA, with subsequent translation into a protein. Large numbers of ncRNAs with biological significance have now been identified, suggesting that ncRNAs are important in their own right and their roles extend far beyond what was originally envisaged. ncRNAs do not only regulate gene expression, but are also involved in chromatin architecture and structural conformation. Several studies have pointed out that ncRNAs participate in heart disease; however, the functions of ncRNAs still remain unclear. ncRNAs are involved in cellular fate, differentiation, proliferation and tissue regeneration, hinting at their potential therapeutic applications. Here, we review the current understanding of both the biological functions and molecular mechanisms of ncRNAs in heart disease and describe some of the ncRNAs that have potential heart regeneration effects.
Collapse
Affiliation(s)
- Oi Kuan Choong
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Desy S Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chen-Yun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Patrick C H Hsieh
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan.,Institute of Medical Genomics and Proteomics, Institute of Clinical Medicine and Department of Surgery, National Taiwan University & Hospital, Taipei 100, Taiwan
| |
Collapse
|
165
|
Lee NY, Chung K, Jin JS, Lee Y, An H. The Inhibitory Effect of Nodakenin on Mast‐Cell‐Mediated Allergic Inflammation Via Downregulation of NF‐κB and Caspase‐1 Activation. J Cell Biochem 2017; 118:3993-4001. [DOI: 10.1002/jcb.26055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/11/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Na Young Lee
- Department of PharmacologyCollege of Korean MedicineSangji UniversityGangwon‐do 220‐702Republic of Korea
- Department of Herbal Medicine ResourcesChonbuk National UniversityIksan 570‐752Republic of Korea
| | - Kyung‐Sook Chung
- Catholic Precision Medicine Research CenterCollege of MedicineThe Catholic University of Korea222, Banpo‐daero, Seocho‐guSeoul 06591Republic of Korea
| | - Jong Sik Jin
- Department of Herbal Medicine ResourcesChonbuk National UniversityIksan 570‐752Republic of Korea
| | - Young‐Cheol Lee
- Department of HerbologyCollege of Korean MedicineSangji UniversityGangwon‐do 220‐702Republic of Korea
| | - Hyo‐Jin An
- Department of PharmacologyCollege of Korean MedicineSangji UniversityGangwon‐do 220‐702Republic of Korea
| |
Collapse
|
166
|
Necroptosis as a potential therapeutic target in multiple organ dysfunction syndrome. Oncotarget 2017; 8:56980-56990. [PMID: 28915647 PMCID: PMC5593618 DOI: 10.18632/oncotarget.18252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 04/17/2017] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate how necroptosisis, i.e. programmed necrosis, is involved in MODS, and to examine whether Nec-1, a specific necroptosis inhibitor, ameliorates multiorgan injury in MODS. Experimental Design A model of MODS was established in six-week old SD rats using fracture trauma followed by hemorrhage. Control animals received sham surgery. Cell death form and necrosome formation were measured by fluorescence-activated cell sorting and western blotting. MODS rats were randomly assigned to receive Nec-1 or saline with pretreatment and once daily. The first end-point was 72 hours survival. Organ injury and dysfunction, inflammatory cytokine levels, and necroptotic execution protein expression were also recorded. Results Organ injury and dysfunction were significantly more severe in the MODS group than the sham group (all p<0.01). Furthermore, MODS-induced liver, lung and kidney tissue injury was characterized by necroptosis rather than apoptosis, and accompanied by necrosome formation. Compared to MODS group, Nec-1 administration significantly improved 72 hours survival (p<0.01). Nec-1 administration significantly reduced necroptosis-induced liver, lung and kidney injury and dysfunction, inhibited inflammatory cytokines production, inhibited release of necroptotic execution proteins such as high-mobility group box 1 and mixed-lineage kinase domain-like protein pseudokinase in MODS rats (all p<0.01). Conclusions These results suggest that necroptosis is involved the pathology of MODS. Further, a necroptotic inhibitor Nec-1 may be considered as an adjunct treatment for MODS.
Collapse
|
167
|
Snyder DT, Hedges JF, Jutila MA. Getting "Inside" Type I IFNs: Type I IFNs in Intracellular Bacterial Infections. J Immunol Res 2017; 2017:9361802. [PMID: 28529959 PMCID: PMC5424489 DOI: 10.1155/2017/9361802] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022] Open
Abstract
Type I interferons represent a unique and complex group of cytokines, serving many purposes during innate and adaptive immunity. Discovered in the context of viral infections, type I IFNs are now known to have myriad effects in infectious and autoimmune disease settings. Type I IFN signaling during bacterial infections is dependent on many factors including whether the infecting bacterium is intracellular or extracellular, as different signaling pathways are activated. As such, the repercussions of type I IFN induction can positively or negatively impact the disease outcome. This review focuses on type I IFN induction and downstream consequences during infection with the following intracellular bacteria: Chlamydia trachomatis, Listeria monocytogenes, Mycobacterium tuberculosis, Salmonella enterica serovar Typhimurium, Francisella tularensis, Brucella abortus, Legionella pneumophila, and Coxiella burnetii. Intracellular bacterial infections are unique because the bacteria must avoid, circumvent, and even co-opt microbial "sensing" mechanisms in order to reside and replicate within a host cell. Furthermore, life inside a host cell makes intracellular bacteria more difficult to target with antibiotics. Because type I IFNs are important immune effectors, modulating this pathway may improve disease outcomes. But first, it is critical to understand the context-dependent effects of the type I IFN pathway in intracellular bacterial infections.
Collapse
Affiliation(s)
- Deann T. Snyder
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Jodi F. Hedges
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Mark A. Jutila
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| |
Collapse
|
168
|
RIPK3 Mediates Necroptosis during Embryonic Development and Postnatal Inflammation in Fadd -Deficient Mice. Cell Rep 2017; 19:798-808. [DOI: 10.1016/j.celrep.2017.04.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/08/2017] [Accepted: 04/03/2017] [Indexed: 01/31/2023] Open
|
169
|
Giricz Z, Koncsos G, Rajtík T, Varga ZV, Baranyai T, Csonka C, Szobi A, Adameová A, Gottlieb RA, Ferdinandy P. Hypercholesterolemia downregulates autophagy in the rat heart. Lipids Health Dis 2017; 16:60. [PMID: 28330474 PMCID: PMC5363032 DOI: 10.1186/s12944-017-0455-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/14/2017] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND We have previously shown that efficiency of ischemic conditioning is diminished in hypercholesterolemia and that autophagy is necessary for cardioprotection. However, it is unknown whether isolated hypercholesterolemia disturbs autophagy or the mammalian target of rapamycin (mTOR) pathways. Therefore, we investigated whether isolated hypercholesterolemia modulates cardiac autophagy-related pathways or programmed cell death mechanisms such as apoptosis and necroptosis in rat heart. METHODS Male Wistar rats were fed either normal chow (NORM; n = 9) or with 2% cholesterol and 0.25% cholic acid-enriched diet (CHOL; n = 9) for 12 weeks. CHOL rats exhibited a 41% increase in plasma total cholesterol level over that of NORM rats (4.09 mmol/L vs. 2.89 mmol/L) at the end of diet period. Animals were sacrificed, hearts were excised and briefly washed out. Left ventricles were snap-frozen for determination of markers of autophagy, mTOR pathway, apoptosis, and necroptosis by Western blot. RESULTS Isolated hypercholesterolemia was associated with a significant reduction in expression of cardiac autophagy markers such as LC3-II, Beclin-1, Rubicon and RAB7 as compared to controls. Phosphorylation of ribosomal S6, a surrogate marker for mTOR activity, was increased in CHOL samples. Cleaved caspase-3, a marker of apoptosis, increased in CHOL hearts, while no difference in the expression of necroptotic marker RIP1, RIP3 and MLKL was detected between treatments. CONCLUSIONS This is the first comprehensive analysis of autophagy and programmed cell death pathways of apoptosis and necroptosis in hearts of hypercholesterolemic rats. Our data show that isolated hypercholesterolemia suppresses basal cardiac autophagy and that the decrease in autophagy may be a result of an activated mTOR pathway. Reduced autophagy was accompanied by increased apoptosis, while cardiac necroptosis was not modulated by isolated hypercholesterolemia. Decreased basal autophagy and elevated apoptosis may be responsible for the loss of cardioprotection reported in hypercholesterolemic animals.
Collapse
Affiliation(s)
- Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Gábor Koncsos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - Tomáš Rajtík
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary
| | - Tamás Baranyai
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - Csaba Csonka
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary
| | - Adrián Szobi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Adriana Adameová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Roberta A. Gottlieb
- Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA 90048 USA
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
170
|
Tran AHV, Han SH, Kim J, Grasso F, Kim IS, Han YS. MutY DNA Glycosylase Protects Cells From Tumor Necrosis Factor Alpha-Induced Necroptosis. J Cell Biochem 2017; 118:1827-1838. [PMID: 28059467 DOI: 10.1002/jcb.25866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/04/2017] [Indexed: 12/27/2022]
Abstract
Numerous studies have implied that mutY DNA glycosylase (MYH) is involved in the repair of post-replicative mispairs and plays a critical role in the base excision repair pathway. Recent in vitro studies have shown that MYH interacts with tumor necrosis factor receptor type 1-associated death domain (TRADD), a key effector protein of tumor necrosis factor receptor-1 (TNFR1) signaling. The association between MYH and TRADD is reversed during tumor necrosis factor alpha (TNF-α)- and camptothecin (CPT)-induced apoptosis, and enhanced during TNF-α-induced survival. After investigating the role of MYH interacts with various proteins following TNF-α stimulation, here, we focus on MYH and TRADD interaction functions in necroptosis and its effects to related proteins. We report that the level of the MYH and TRADD complex was also reduced during necroptosis induced by TNF-α and zVAD-fmk. In particular, we also found that MYH is a biologically important necrosis suppressor. Under combined TNF-α and zVAD-fmk treatment, MYH-deficient cells were induced to enter the necroptosis pathway but primary mouse embryonic fibroblasts (MEFs) were not. Necroptosis in the absence of MYH proceeds via the inactivation of caspase-8, followed by an increase in the formation of the kinase receptor- interacting protein 1 (RIP1)-RIP3 complex. Our results suggested that MYH, which interacts with TRADD, inhibits TNF-α necroptotic signaling. Therefore, MYH inactivation is essential for necroptosis via the downregulation of caspase-8. J. Cell. Biochem. 118: 1827-1838, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- An Hue Vy Tran
- Department of Advanced Technology Fusion, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Korea
| | - Se Hee Han
- Department of Advanced Technology Fusion, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea
| | - Joon Kim
- Laboratory of Biochemistry, School of Life Sciences and Biotechnology and BioInstitute, Korea University, Seoul, Korea
| | - Francesca Grasso
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Rome, Lazio, Italy
| | - In San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Korea
| | - Ye Sun Han
- Department of Advanced Technology Fusion, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Korea
| |
Collapse
|
171
|
Kong D, Zhu J, Liu Q, Jiang Y, Xu L, Luo N, Zhao Z, Zhai Q, Zhang H, Zhu M, Liu X. Mesenchymal stem cells protect neurons against hypoxic-ischemic injury via inhibiting parthanatos, necroptosis, and apoptosis, but not autophagy. Cell Mol Neurobiol 2017; 37:303-313. [PMID: 27044018 PMCID: PMC11482119 DOI: 10.1007/s10571-016-0370-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/24/2016] [Indexed: 01/01/2023]
Abstract
Cellular therapy with mesenchymal stem cells (MSCs) protects cortical neurons against hypoxic-ischemic injury of stroke. Although sorts of efforts have been made to confirm the neuroprotective effect of MSCs on neurons against hypoxic-ischemic injury, the mechanism is until now far away from clear. Here in this study, oxygen-glucose deprivation (OGD)-injured neuron model was applied to mimic the neuronal hypoxic-ischemic injury in vitro. Co-culturing with MSCs in a transwell co-culture system, the OGD injured neurons were rescued by 75.0 %. Further data demonstrated that co-culturing with MSCs protected the cortical neurons from the OGD-induced parthanatos by alleviating apoptosis-inducing factor (AIF) nuclear translocation; attenuated the neuronal necroptosis by down-regulating the expression of the two essential kinases in necroptosis, receptor interacting protein kinase1 (RIP1) and 3 (RIP3); rescued the neurons from apoptosis by deactivating caspase-3; whilst performed no significant influence on OGD-induced neuronal autophagy, according to its failed regulation on Beclin1. In conclusion, MSCs potentially protect the cortical neurons from OGD-injury in vitro, through rescuing neurons from the cell death of parthanatos, necroptosis, and apoptosis, but not autophagy, which could provide some evidence to the mechanism explanation on stem cell treatment for ischemic stroke.
Collapse
Affiliation(s)
- Deyan Kong
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
- Department of Neurology, The Affiliated Ruikang Hospital of Guangxi Traditional Chinese Medical University, Nanning, 530011, Guangxi Zhuang Autonomous Region, China
| | - Juehua Zhu
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Qian Liu
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Yongjun Jiang
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Lily Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Ning Luo
- Department of Neurology, The Affiliated Ruikang Hospital of Guangxi Traditional Chinese Medical University, Nanning, 530011, Guangxi Zhuang Autonomous Region, China
| | - Zhenqiang Zhao
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
- Department of Neurology, Affiliated Hospital of Hainan Medical College, Haikou, 570102, Hainan, China
| | - Qijin Zhai
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Hao Zhang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Mingyue Zhu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
| | - Xinfeng Liu
- Department of Neurology, Jinling Hospital, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
172
|
Peddireddy V, Doddam SN, Ahmed N. Mycobacterial Dormancy Systems and Host Responses in Tuberculosis. Front Immunol 2017; 8:84. [PMID: 28261197 PMCID: PMC5309233 DOI: 10.3389/fimmu.2017.00084] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/18/2017] [Indexed: 12/15/2022] Open
Abstract
Tuberculosis (TB) caused by the intracellular pathogen, Mycobacterium tuberculosis (Mtb), claims more than 1.5 million lives worldwide annually. Despite promulgation of multipronged strategies to prevent and control TB, there is no significant downfall occurring in the number of new cases, and adding to this is the relapse of the disease due to the emergence of antibiotic resistance and the ability of Mtb to remain dormant after primary infection. The pathology of Mtb is complex and largely attributed to immune-evading strategies that this pathogen adopts to establish primary infection, its persistence in the host, and reactivation of pathogenicity under favorable conditions. In this review, we present various biochemical, immunological, and genetic strategies unleashed by Mtb inside the host for its survival. The bacterium enables itself to establish a niche by evading immune recognition via resorting to masking, establishment of dormancy by manipulating immune receptor responses, altering innate immune cell fate, enhancing granuloma formation, and developing antibiotic tolerance. Besides these, the regulatory entities, such as DosR and its regulon, encompassing various putative effector proteins play a vital role in maintaining the dormant nature of this pathogen. Further, reactivation of Mtb allows relapse of the disease and is favored by the genes of the Rtf family and the conditions that suppress the immune system of the host. Identification of target genes and characterizing the function of their respective antigens involved in primary infection, dormancy, and reactivation would likely provide vital clues to design novel drugs and/or vaccines for the control of dormant TB.
Collapse
Affiliation(s)
- Vidyullatha Peddireddy
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad , Hyderabad , India
| | - Sankara Narayana Doddam
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad , Hyderabad , India
| | - Niyaz Ahmed
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India; Laboratory Sciences and Services Division, International Centre for Diarrhoeal Disease Research Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
173
|
The MLKL Channel in Necroptosis Is an Octamer Formed by Tetramers in a Dyadic Process. Mol Cell Biol 2017; 37:MCB.00497-16. [PMID: 27920255 DOI: 10.1128/mcb.00497-16] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/28/2016] [Indexed: 12/18/2022] Open
Abstract
Oligomerization of the mixed-lineage kinase domain-like protein (MLKL) is essential for its cation channel function in necroptosis. Here we show that the MLKL channel is an octamer comprising two previously identified tetramers most likely in their side-by-side position. Intermolecule disulfide bonds are present in the tetramer but are not required for octamer assembly and necroptosis. MLKL forms oligomers in the necrosome and is then released from the necrosome before or during its membrane translocation. We identified two MLKL mutants that could not oligomerize into octamers, although they formed a tetramer, and also, one MLKL mutant could spontaneously form a disulfide bond-linked octamer. Subsequent analysis revealed that the tetramers fail to translocate to the plasma membrane and that the MLKL octamer formation depends on α-helices 4 and 5. While MLKL could be detected from outside the cells, its N- and C-terminal ends could not be detected, indicating that the MLKL octamer spans across the plasma membrane, leaving its N and C termini inside the cell. These data allowed us to propose a 180° symmetry model of the MLKL octamer and conclude that the fully assembled MLKL octamers, but not the previously described tetramers, act as effectors of necroptosis.
Collapse
|
174
|
Ramachandran A, Jaeschke H. Mechanisms of acetaminophen hepatotoxicity and their translation to the human pathophysiology. J Clin Transl Res 2017; 3:157-169. [PMID: 28670625 PMCID: PMC5489132 DOI: 10.18053/jctres.03.2017s1.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 12/15/2022] Open
Abstract
Acetaminophen (APAP) overdose is the most common cause of acute liver failure in the United States and mechanisms of liver injury induced by APAP overdose have been the focus of extensive investigation. Studies in the mouse model, which closely reproduces the human condition, have shown that hepatotoxicity is initiated by formation of a reactive metabolite N-acetyl-p-benzoquinone imine (NAPQI), which depletes cellular glutathione and forms protein adducts on mitochondrial proteins. This leads to mitochondrial oxidative and nitrosative stress, accompanied by activation of c-jun N-terminal kinase (JNK) and its translocation to the mitochondria. This then amplifies the mitochondrial oxidant stress, resulting in translocation of Bax and dynamin related protein 1 (Drp1) to the mitochondria, which induces mitochondrial fission, and ultimately induction of the mitochondrial membrane permeability transition (MPT). The induction of MPT triggers release of intermembrane proteins such as apoptosis inducing factor (AIF) and endonuclease G into the cytosol and their translocation to the nucleus, causing nuclear DNA fragmentation and activation of regulated necrosis. Though these cascades of events were primarily identified in the mouse model, studies on human hepatocytes and analysis of circulating biomarkers from patients after APAP overdose, indicate that a number of mechanistic events are identical in mice and humans. Circulating biomarkers also seem to be useful in predicting the course of liver injury after APAP overdose in humans and hold promise for significant clinical use in the near future.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
175
|
Abstract
Drug-induced hepatotoxicity (DIH) is a significant cause of acute liver failure and liver transplantation. Diagnosis is challenging due to the idiosyncratic nature, its presentation in the form of other liver disease, and the lack of a definite diagnostic criteria. Generation of reactive metabolites, oxidative stress, and mitochondrial dysfunction are common mechanisms involved in DIH. Certain risk factors associated with a drug and within an individual further predispose patients to DIH.
Collapse
Affiliation(s)
- Amina Ibrahim Shehu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 3rd Floor Salk Pavillion, Pittsburgh, PA 15261, USA
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 3rd Floor Salk Pavillion, Pittsburgh, PA 15261, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, 718 Salk Hall, 3501 Terrace Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
176
|
Tert-butyl hydroperoxide (t-BHP) induced apoptosis and necroptosis in endothelial cells: Roles of NOX4 and mitochondrion. Redox Biol 2017; 11:524-534. [PMID: 28088644 PMCID: PMC5237803 DOI: 10.1016/j.redox.2016.12.036] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/25/2016] [Accepted: 12/22/2016] [Indexed: 10/25/2022] Open
Abstract
Oxidative stress causes endothelial death while underlying mechanisms remain elusive. Herein, the pro-death effect of tert-butyl hydroperoxide (t-BHP) was investigated with low concentration (50μM) of t-BHP (t-BHPL) and high concentration (500μM) of t-BHP (t-BHPH). Both t-BHPL and t-BHPH induced endothelial cell death was determined. T-BHPL induced caspase-dependent apoptosis and reactive oxygen species (ROS) generation, which was inhibited by N-acetyl-L-cysteine (NAC). Furthermore, NADPH oxidase inhibitor diphenyleneiodonium (DPI), NOX4 siRNA, and NOX4 inhibitor GKT137831 reduced t-BHPL-induced ROS generation while mitochondrial respiratory chain inhibitors rotenone (Rot), 2-thenoyltrifluoroacetone (TTFA), and antimycin A (AA) failed to do so. NOX4 overexpression resulted in increased ROS generation and Akt expression but decreased sensitivity to t-BHPL. In contrast, T-BHPH induced LDH release, PI uptake, and cell translucent cytoplasm. RIP1 inhibitor necrostatin-1 (Nec-1), MLKL inhibitor necrosulfonamide (NSA) and silencing RIP1, RIP3, and MLKL inhibited t-BHPH-induced cell death while pan-caspase inhibitor Z-VAD-FMK showed no effect. T-BHPH-induced ROS production was inhibited by TTFA, AA and Rot while DPI showed no effect. T-BHPH induced RIP1/RIP3 interaction, which was decreased by Rot, TTFA, and AA. Silence RIP1 and RIP3 but not MLKL inhibited t-BHPH-induced mitochondrial membrane potential (MMP) decrease and ROS production. Moreover, P38MAPK inhibitor SB203580 reversed both t-BHPL and t-BHPH-induced cell death while inhibitors for ERKs and JNKs showed no obvious effect. These data suggested that t-BHP induced both apoptosis and necroptosis in endothelial cells which was mediated by ROS and p38MAPK. ROS derived from NADPH oxidase and mitochondria contributed to t-BHPL and t-BHPH-induced apoptosis and necroptosis, respectively.
Collapse
|
177
|
Chao X, Wang S, Ding WX. Cell Death in Alcohol-Induced Liver Injury. CELLULAR INJURY IN LIVER DISEASES 2017:119-142. [DOI: 10.1007/978-3-319-53774-0_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
178
|
Cancer's Achilles' Heel: Apoptosis and Necroptosis to the Rescue. Int J Mol Sci 2016; 18:ijms18010023. [PMID: 28025559 PMCID: PMC5297658 DOI: 10.3390/ijms18010023] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/05/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022] Open
Abstract
Apoptosis, and the more recently discovered necroptosis, are two avenues of programmed cell death. Cancer cells survive by evading these two programs, driven by oncogenes and tumor suppressor genes. While traditional therapy using small molecular inhibitors and chemotherapy are continuously being utilized, a new and exciting approach is actively underway by identifying and using synergistic relationship between driver and rescue genes in a cancer cell. Through these synthetic lethal relationships, we are gaining tremendous insights into tumor vulnerabilities and specific molecular avenues for induction of programmed cell death. In this review, we briefly discuss the two cell death processes and cite examples of such synergistic manipulations for therapeutic purposes.
Collapse
|
179
|
Galluzzi L, Kepp O, Chan FKM, Kroemer G. Necroptosis: Mechanisms and Relevance to Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 12:103-130. [PMID: 27959630 DOI: 10.1146/annurev-pathol-052016-100247] [Citation(s) in RCA: 513] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Necroptosis is a form of regulated cell death that critically depends on receptor-interacting serine-threonine kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) and generally manifests with morphological features of necrosis. The molecular mechanisms that underlie distinct instances of necroptosis have just begun to emerge. Nonetheless, it has already been shown that necroptosis contributes to cellular demise in various pathophysiological conditions, including viral infection, acute kidney injury, and cardiac ischemia/reperfusion. Moreover, human tumors appear to obtain an advantage from the downregulation of key components of the molecular machinery for necroptosis. Although such an advantage may stem from an increased resistance to adverse microenvironmental conditions, accumulating evidence indicates that necroptosis-deficient cancer cells are poorly immunogenic and hence escape natural and therapy-elicited immunosurveillance. Here, we discuss the molecular mechanisms and relevance to disease of necroptosis.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065; .,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; .,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Oliver Kepp
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; .,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France;
| | | | - Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; .,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France; .,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, 17176 Stockholm, Sweden.,Pôle de Biologie, Hôpital Européen George Pompidou, AP-HP, 75015 Paris, France
| |
Collapse
|
180
|
Ma B, Marcotte D, Paramasivam M, Michelsen K, Wang T, Bertolotti-Ciarlet A, Jones JH, Moree B, Butko M, Salafsky J, Sun X, McKee T, Silvian LF. ATP-Competitive MLKL Binders Have No Functional Impact on Necroptosis. PLoS One 2016; 11:e0165983. [PMID: 27832137 PMCID: PMC5104457 DOI: 10.1371/journal.pone.0165983] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/20/2016] [Indexed: 01/18/2023] Open
Abstract
MLKL is a pore forming pseudokinase involved in the final stage of necroptosis, a form of programmed cell death. Its phosphorylation by RIPK3 is necessary for triggering necroptosis but not for triggering apoptosis, which makes it a unique target for pharmacological inhibition to block necroptotic cell death. This mechanism has been described as playing a role in disease progression in neurodegenerative and inflammatory diseases. A type II kinase inhibitor (cpd 1) has been described that reportedly binds to the MLKL pseudokinase domain and prevents necroptosis. Here we describe five compounds that bind to the MLKL ATP-binding site, however the four MLKL-selective compounds have no activity in rescuing cells from necroptosis. We use kinase selectivity panels, crystallography and a new conformationally sensitive method of measuring protein conformational changes (SHG) to confirm that the one previously reported compound that can rescue cells (cpd 1) is a non-selective type II inhibitor that also inhibits the upstream kinase RIPK1. Although this compound can shift the GFE motif of the activation loop to an “out” position, the accessibility of the key residue Ser358 in the MLKL activation loop is not affected by compound binding to the MLKL active site. Our studies indicate that an ATP-pocket inhibitor of the MLKL pseudokinase domain does not have any impact on the necroptosis pathway, which is contrary to a previously reported study.
Collapse
Affiliation(s)
- Bin Ma
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | - Doug Marcotte
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | | | - Klaus Michelsen
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | - Ti Wang
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | | | - John Howard Jones
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | - Ben Moree
- Biodesy Inc., South San Francisco, CA, 94080, United States of America
| | - Margaret Butko
- Biodesy Inc., South San Francisco, CA, 94080, United States of America
| | - Joshua Salafsky
- Biodesy Inc., South San Francisco, CA, 94080, United States of America
| | - Xin Sun
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | - Timothy McKee
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
| | - Laura F. Silvian
- Drug Discovery, Biogen Inc., Cambridge, MA, 02142, United States of America
- * E-mail:
| |
Collapse
|
181
|
Dong W, Zhou M, Dong M, Pan B, Liu Y, Shao J, Gu X, Huang Y, Li G, Wang Y, Sun H. Keto acid metabolites of branched-chain amino acids inhibit oxidative stress-induced necrosis and attenuate myocardial ischemia-reperfusion injury. J Mol Cell Cardiol 2016; 101:90-98. [PMID: 27832938 DOI: 10.1016/j.yjmcc.2016.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 01/06/2023]
Abstract
Branched chain α-keto acids (BCKAs) are endogenous metabolites of branched-chain amino acids (BCAAs). BCAA and BCKA are significantly elevated in pathologically stressed heart and contribute to chronic pathological remodeling and dysfunction. However, their direct impact on acute cardiac injury is unknown. Here, we demonstrated that elevated BCKAs significantly attenuated ischemia-reperfusion (I/R) injury and preserved post I/R function in isolated mouse hearts. BCKAs protected cardiomyocytes from oxidative stress-induced cell death in vitro. Mechanistically, BCKA protected oxidative stress induced cell death by inhibiting necrosis without affecting apoptosis or autophagy. Furthermore, BCKAs, but not BCAAs, protected mitochondria and energy production from oxidative injury. Finally, administration of BCKAs during reperfusion was sufficient to significantly attenuate cardiac I/R injury. These findings uncover an unexpected role of BCAA metabolites in cardioprotection against acute ischemia/reperfusion injury, and demonstrate the potential use of BCKA treatment to preserve ischemic tissue during reperfusion.
Collapse
Affiliation(s)
- Weibing Dong
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meiyi Zhou
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mei Dong
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Bangfen Pan
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunxia Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Shao
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoping Gu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Huang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yibin Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Division of Molecular Medicine, Departments of Anesthesiology, Medicine and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Haipeng Sun
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Division of Molecular Medicine, Departments of Anesthesiology, Medicine and Physiology, Molecular Biology Institute, Cardiovascular Research Laboratories, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
182
|
Ornelas A, McCullough CR, Lu Z, Zacharias NM, Kelderhouse LE, Gray J, Yang H, Engel BJ, Wang Y, Mao W, Sutton MN, Bhattacharya PK, Bast RC, Millward SW. Induction of autophagy by ARHI (DIRAS3) alters fundamental metabolic pathways in ovarian cancer models. BMC Cancer 2016; 16:824. [PMID: 27784287 PMCID: PMC5080741 DOI: 10.1186/s12885-016-2850-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/10/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Autophagy is a bulk catabolic process that modulates tumorigenesis, therapeutic resistance, and dormancy. The tumor suppressor ARHI (DIRAS3) is a potent inducer of autophagy and its expression results in necroptotic cell death in vitro and tumor dormancy in vivo. ARHI is down-regulated or lost in over 60 % of primary ovarian tumors yet is dramatically up-regulated in metastatic disease. The metabolic changes that occur during ARHI induction and their role in modulating death and dormancy are unknown. METHODS We employed Nuclear Magnetic Resonance (NMR)-based metabolomic strategies to characterize changes in key metabolic pathways in both cell culture and xenograft models of ARHI expression and autophagy. These pathways were further interrogated by cell-based immunofluorescence imaging, tracer uptake studies, targeted metabolic inhibition, and in vivo PET/CT imaging. RESULTS Induction of ARHI in cell culture models resulted in an autophagy-dependent increase in lactate production along with increased glucose uptake and enhanced sensitivity to glycolytic inhibitors. Increased uptake of glutamine was also dependent on autophagy and dramatically sensitized cultured ARHI-expressing ovarian cancer cell lines to glutaminase inhibition. Induction of ARHI resulted in a reduction in mitochondrial respiration, decreased mitochondrial membrane potential, and decreased Tom20 staining suggesting an ARHI-dependent loss of mitochondrial function. ARHI induction in mouse xenograft models resulted in an increase in free amino acids, a transient increase in [18F]-FDG uptake, and significantly altered choline metabolism. CONCLUSIONS ARHI expression has previously been shown to trigger autophagy-associated necroptosis in cell culture. In this study, we have demonstrated that ARHI expression results in decreased cellular ATP/ADP, increased oxidative stress, and decreased mitochondrial function. While this bioenergetic shock is consistent with programmed necrosis, our data indicates that the accompanying up-regulation of glycolysis and glutaminolysis is autophagy-dependent and serves to support cell viability rather than facilitate necroptotic cell death. While the mechanistic basis for metabolic up-regulation following ARHI induction is unknown, our preliminary data suggest that decreased mitochondrial function and increased metabolic demand may play a role. These alterations in fundamental metabolic pathways during autophagy-associated necroptosis may provide the basis for new therapeutic strategies for the treatment of dormant ovarian tumors.
Collapse
Affiliation(s)
- Argentina Ornelas
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Christopher R McCullough
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Zhen Lu
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Niki M Zacharias
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA.,Department of Bioengineering, Rice University, Houston, USA
| | - Lindsay E Kelderhouse
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Joshua Gray
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Hailing Yang
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Brian J Engel
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Yan Wang
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Weiqun Mao
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Margie N Sutton
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Pratip K Bhattacharya
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Robert C Bast
- Department of Experimental Therapeutics, the University of Texas M.D. Anderson Cancer Center, Houston, USA
| | - Steven W Millward
- Department of Cancer Systems Imaging, the University of Texas M.D. Anderson Cancer Center, Houston, USA. .,Department of Bioengineering, Rice University, Houston, USA.
| |
Collapse
|
183
|
Yang DW, Kang OH, Lee YS, Han SH, Lee SW, Cha SW, Seo YS, Mun SH, Gong R, Shin DW, Kwon DY. Anti-inflammatory effect of salidroside on phorbol-12-myristate-13-acetate plus A23187-mediated inflammation in HMC-1 cells. Int J Mol Med 2016; 38:1864-1870. [PMID: 27779653 DOI: 10.3892/ijmm.2016.2781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/27/2016] [Indexed: 11/05/2022] Open
Abstract
Salidroside [2-(4-hydroxyphenyl)ethyl β-D-gluco-pyranoside (SAS)] has been identified as the most potent ingredient of the plant Rhodiola rosea L. Previous studies have demonstrated that it possesses a number of pharmacological properties, including anti-aging, anti-fatigue, antioxidant, anticancer and anti-inflammatory properties. In this study, to ascertain the molecular mechanisms responsible for the anti-inflammatory activity of SAS, we used phorbol-12-myristate-13-acetate (PMA) plus A23187 to induce inflammation in human mast cell line-1 (HMC-1). The HMC-1 cells were treated with SAS prior to being stimulated with PMA plus A23187. Pro-inflammatory cytokine production was measured by enzyme-linked immunosorbent assay (ELISA) and reverse transcription-polymerase chain reaction (RT-PCR). Western blot analysis was used to examine the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB). SAS inhibited the mRNA expression and production of interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF). In cells stimulated with PMA plus A23187, SAS suppressed the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and c-jun N-terminal kinase 1/2 (JNK1/2), but not that of p38 MAPK. SAS suppressed the expression of NF-κB in the nucleus. On the whole, our results suggest that SAS exerts an anti-inflammatory effect by inhibiting the production of pro-inflammatory cytokines through the blocking of the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Da-Wun Yang
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ok-Hwa Kang
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Sin-Hee Han
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Sang-Won Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Seon-Woo Cha
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, Chungbuk 369-873, Republic of Korea
| | - Yun-Soo Seo
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | - Su-Hyun Mun
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Ryong Gong
- BK21 Plus Team, Professional Graduate School of Oriental Medicine,Wonkwang University, Iksan, Jeonbuk 570‑749, Republic of Korea
| | - Dong-Won Shin
- Department of Oriental Medicine Resources, Sunchon National University, Jeonnam 540-742, Republic of Korea
| | - Dong-Yeul Kwon
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Institute of Biotechnology, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| |
Collapse
|
184
|
Cui H, Zhu Y, Yang Q, Zhao W, Zhang S, Zhou A, Jiang D. Necrostatin-1 treatment inhibits osteocyte necroptosis and trabecular deterioration in ovariectomized rats. Sci Rep 2016; 6:33803. [PMID: 27703177 PMCID: PMC5050438 DOI: 10.1038/srep33803] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022] Open
Abstract
Estrogen (E2) deficiency has been associated with accelerated osteocyte apoptosis. Our previous study showed necroptosis accelerated the loss of osteocytes in E2 deficiency-induced osteoporosis in rats in addition to apoptosis, but the mechanism involved remains. Necroptosis is a caspase-independent form of programmed cell death. In the necroptosis pathway, receptor interaction proteins 1 and 3 (RIP1/3) play vital roles. Necrostatin-1 (Nec-1) has been confirmed to be a specific inhibitor of necroptosis. However, the effect of Nec-1 on postmenopausal osteoporosis remains ambiguous. The aim of this study was to investigate the effect of Nec-1 on osteocytes in ovariectomized (OVX) rats. We found that an increased number of necroptotic osteocytes was related to the production of tumor necrosis factor-alpha (TNF-α) in OVX rats. Treatment with Nec-1 significantly decreased RIP1 and RIP3 expression in OVX rats and inhibited osteocyte necroptosis induced by TNF-α in vitro. Both E2 and Nec-1 treatment markedly ameliorated trabecular bone deterioration. Nec-1 also significantly elevated the levels of bone formation markers and decreased bone resorption markers. These data suggest that the role of Nec-1 on alleviating bone loss might be associated with Nec-1 restraining TNF-α-induced osteocyte necroptosis in rats with E2 deficiency-induced osteoporosis. This process may represent a novel therapeutic strategy for the treatment of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Hongwang Cui
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yongjun Zhu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiming Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weikang Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiyang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dianming Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
185
|
Koppe C, Verheugd P, Gautheron J, Reisinger F, Kreggenwinkel K, Roderburg C, Quagliata L, Terracciano L, Gassler N, Tolba RH, Boege Y, Weber A, Karin M, Luedde M, Neumann UP, Weiskirchen R, Tacke F, Vucur M, Trautwein C, Lüscher B, Preisinger C, Heikenwalder M, Luedde T. IκB kinaseα/β control biliary homeostasis and hepatocarcinogenesis in mice by phosphorylating the cell-death mediator receptor-interacting protein kinase 1. Hepatology 2016; 64:1217-1231. [PMID: 27396433 DOI: 10.1002/hep.28723] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 07/01/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED The IκB-Kinase (IKK) complex-consisting of the catalytic subunits, IKKα and IKKβ, as well as the regulatory subunit, NEMO-mediates activation of the nuclear factor κB (NF-κB) pathway, but previous studies suggested the existence of NF-κB-independent functions of IKK subunits with potential impact on liver physiology and disease. Programmed cell death is a crucial factor in the progression of liver diseases, and receptor-interacting kinases (RIPKs) exerts strategic control over multiple pathways involved in regulating novel programmed cell-death pathways and inflammation. We hypothesized that RIPKs might be unrecognized targets of the catalytic IKK-complex subunits, thereby regulating hepatocarcinogenesis and cholestasis. In this present study, mice with specific genetic inhibition of catalytic IKK activity in liver parenchymal cells (LPCs; IKKα/β(LPC-KO) ) were intercrossed with RIPK1(LPC-KO) or RIPK3(-/-) mice to examine whether RIPK1 or RIPK3 might be downstream targets of IKKs. Moreover, we performed in vivo phospho-proteome analyses and in vitro kinase assays, mass spectrometry, and mutagenesis experiments. These analyses revealed that IKKα and IKKβ-in addition to their known function in NF-κB activation-directly phosphorylate RIPK1 at distinct regions of the protein, thereby regulating cell viability. Loss of this IKKα/β-dependent RIPK1 phosphorylation in LPCs inhibits compensatory proliferation of hepatocytes and intrahepatic biliary cells, thus impeding HCC development, but promoting biliary cell paucity and lethal cholestasis. CONCLUSIONS IKK-complex subunits transmit a previously unrecognized signal through RIPK1, which is fundamental for the long-term consequences of chronic hepatic inflammation and might have potential implications for future pharmacological strategies against cholestatic liver disease and cancer. (Hepatology 2016;64:1217-1231).
Collapse
Affiliation(s)
- Christiane Koppe
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Patricia Verheugd
- Institute of Biochemistry and Molecular Biology, University Hospital RWTH Aachen University, Aachen, Germany
| | - Jérémie Gautheron
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Florian Reisinger
- Department of Virology, Technische Universität München/Helmholtz Zentrum München für Gesundheit und Umwelt (HMGU), Munich, Germany
| | - Karina Kreggenwinkel
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christoph Roderburg
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Luca Quagliata
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Luigi Terracciano
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Nikolaus Gassler
- Institute of Pathology, Hospital of Braunschweig, Braunschweig, Germany
| | - René H Tolba
- Department of Laboratory Animal Research, University Hospital RWTH Aachen, Aachen, Germany
| | - Yannick Boege
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Achim Weber
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Karin
- Department of Pharmacology and Pathology, School of Medicine, UCSD, La Jolla, CA
| | - Mark Luedde
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ulf P Neumann
- Department of Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Mihael Vucur
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, University Hospital RWTH Aachen University, Aachen, Germany
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Center for Clinical Research (IZKF) Aachen, University Hospital RWTH Aachen, Aachen, Germany
| | - Mathias Heikenwalder
- Department of Virology, Technische Universität München/Helmholtz Zentrum München für Gesundheit und Umwelt (HMGU), Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tom Luedde
- Department of Medicine III, Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
186
|
Yamaguchi N, Sekine S, Naguro I, Sekine Y, Ichijo H. KLHDC10 Deficiency Protects Mice against TNFα-Induced Systemic Inflammation. PLoS One 2016; 11:e0163118. [PMID: 27631783 PMCID: PMC5025154 DOI: 10.1371/journal.pone.0163118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/03/2016] [Indexed: 12/30/2022] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is a form of fatal acute inflammation for which there is no effective treatment. Here, we revealed that the ablation of Kelch domain containing 10 (KLHDC10), which we had originally identified as an activator of Apoptosis Signal-regulating Kinase 1 (ASK1), protects mice against TNFα-induced SIRS. The disease development of SIRS is mainly divided into two stages. The early stage is characterized by TNFα-induced systemic necroptosis, a regulated form of necrosis mediated by Receptor-interacting protein (RIP) 1/3 kinases. The later stage presents with an over-production of inflammatory cytokines induced by damage-associated molecular patterns (DAMPs), which are immunogenic cellular contents released from cells that underwent necroptosis. Analysis of TNFα-challenged mice revealed that KLHDC10-deficient mice show a reduction in the inflammatory response, but not in early systemic necroptosis. In vitro analysis suggested that the reduced inflammatory response observed in KLHDC10-deficient mice might be caused, in part, by enhanced necroptosis of inflammatory cells encountering DAMPs. Interestingly, the enhancement of necroptosis induced by KLHDC10 deficiency was selectively observed in inflammatory cells. Our results suggest that KLHDC10 is a cell-type specific regulator of necroptosis that ultimately contributes to the development of TNFα-induced SIRS.
Collapse
Affiliation(s)
- Namiko Yamaguchi
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Shiori Sekine
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Isao Naguro
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yusuke Sekine
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
187
|
Mao Y, Chen X, Xu M, Fujita K, Motoki K, Sasabe T, Homma H, Murata M, Tagawa K, Tamura T, Kaye J, Finkbeiner S, Blandino G, Sudol M, Okazawa H. Targeting TEAD/YAP-transcription-dependent necrosis, TRIAD, ameliorates Huntington’s disease pathology. Hum Mol Genet 2016; 25:4749-4770. [DOI: 10.1093/hmg/ddw303] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/20/2016] [Accepted: 08/26/2016] [Indexed: 11/14/2022] Open
|
188
|
Caspase Inhibition Prevents Tumor Necrosis Factor-α-Induced Apoptosis and Promotes Necrotic Cell Death in Mouse Hepatocytes in Vivo and in Vitro. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2623-36. [PMID: 27616656 DOI: 10.1016/j.ajpath.2016.06.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/24/2016] [Accepted: 06/09/2016] [Indexed: 02/07/2023]
Abstract
How different cell death modes and cell survival pathways cross talk remains elusive. We determined the interrelation of apoptosis, necrosis, and autophagy in tumor necrosis factor (TNF)-α/actinomycin D (ActD) and lipopolysaccharide/D-galactosamine (GalN)-induced hepatotoxicity in vitro and in vivo. We found that TNF-α/ActD-induced apoptosis was completely blocked by a general caspase inhibitor ZVAD-fmk at 24 hours but hepatocytes still died by necrosis at 48 hours. Inhibition of caspases also protected mice against lipopolysaccharide/GalN-induced apoptosis and liver injury at the early time point, but this protection was diminished after prolonged treatment by switching apoptosis to necrosis. Inhibition of receptor-interacting protein kinase (RIP)1 by necrostatin 1 partially inhibited TNF-α/ZVAD-induced necrosis in primary hepatocytes. Pharmacologic inhibition of autophagy or genetic deletion of Atg5 in hepatocytes did not protect against TNF-α/ActD/ZVAD-induced necrosis. Moreover, pharmacologic inhibition of RIP1 or genetic deletion of RIP3 failed to protect and even exacerbated liver injury after mice were treated with lipopolysaccharide/GalN and a pan-caspase inhibitor. In conclusion, our results suggest that different cell death mode and cell survival pathways are closely integrated during TNF-α-induced liver injury when both caspases and NF-κB are blocked. Moreover, results from our study also raised concerns about the safety of currently ongoing clinical trials that use caspase inhibitors.
Collapse
|
189
|
Pan L, Yao DC, Yu YZ, Li SJ, Chen BJ, Hu GH, Xi C, Wang ZH, Wang HY, Li JH, Tu YS. Necrostatin-1 protects against oleic acid-induced acute respiratory distress syndrome in rats. Biochem Biophys Res Commun 2016; 478:1602-8. [PMID: 27586277 DOI: 10.1016/j.bbrc.2016.08.163] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/29/2016] [Indexed: 01/03/2023]
Abstract
Necroptosis is a recently discovered necrotic cell death which is regulated by receptor interacting protein kinase 1 (RIPK1) and RIPK3 under the stimulus of death signal and can be inhibited by necrostatin-1 (Nec-1) specifically. Therefore, the aim was to investigate the role of necroptosis in a rat model of acute respiratory distress syndrome (ARDS) induced by oleic acid (OA) and assess the effect of Nec-1 on lung injury in ARDS. Our results found that RIPK1, RIPK3 and mixed lineage kinase domain-like protein (MLKL) were abundantly expressed in rat lung tissues of OA-induced ARDS. Nec-1 pretreatment improved pulmonary function and attenuated lung edema dramatically in OA-induced ARDS rats. Furthermore, Nec-1 reduced RIPK1-RIPK3 interaction and down-regulated RIPK1-RIPK3-MLKL signal pathway, and inhibited inflammatory response by reducing neutrophil infiltration and protein leakage into lung tissue in OA-induced ARDS. Collectively, our study proves the intervention of necroptosis in OA-induced ARDS. Moreover, our findings imply that Nec-1 plays an important role in the treatment of ARDS via inhibiting necroptosis and inflammation.
Collapse
Affiliation(s)
- Long Pan
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Dun-Chen Yao
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Yu-Zhong Yu
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Sheng-Jie Li
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The First Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Bing-Jun Chen
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Gui-He Hu
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Chang Xi
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Third Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Zi-Hui Wang
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China; The Second Clinical Medical College, Guangzhou Medical University, Guangzhou 511436, China
| | - Hong-Yan Wang
- Department of Pathology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jian-Hua Li
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yong-Sheng Tu
- Department of Physiology, School of Basic Sciences, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
190
|
Diao Y, Ma X, Min W, Lin S, Kang H, Dai Z, Wang X, Zhao Y. Dasatinib promotes paclitaxel-induced necroptosis in lung adenocarcinoma with phosphorylated caspase-8 by c-Src. Cancer Lett 2016; 379:12-23. [PMID: 27195913 DOI: 10.1016/j.canlet.2016.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/09/2016] [Accepted: 05/03/2016] [Indexed: 12/25/2022]
Abstract
Cisplatin and paclitaxel are considered to be the backbone of chemotherapy in lung adenocarcinoma. These agents show pleiotropic effects on cell death. However, the precise mechanisms remain unclear. The present study reported that phosphorylated caspase-8 at tyrosine 380 (p-Casp8) was characterized as a biomarker of chemoresistance to TP regimen (cisplatin and paclitaxel) in patients with resectable lung adenocarcinoma with significantly poorer 5-year disease-free survival (DFS) and overall survival (OS). Cisplatin killed lung adenocarcinoma cells regardless of c-Src-induced caspase-8 phosphorylation at tyrosine 380. Subsequently, we identified a novel mechanism by which paclitaxel induced necroptosis in lung adenocarcinoma cells that was dependent upon p-Casp8, receptor-interacting protein kinase 1 (RIPK1), and RIPK3. Moreover, dasatinib, a c-Src inhibitor, dephosphorylated caspase-8 to facilitate necroptosis, rather than apoptosis, in paclitaxel-treated p-Casp8-expressing lung adenocarcinoma cells. The data from our study revealed previously unrecognized roles of p-Casp8 as a positive effector in the initiation of necroptosis and as a negative effector in the repression of the interaction between RIPK1 and RIPK3. Moreover, these outcomes supported the need for further clinical studies with the goal of evaluating the efficacy of dasatinib plus paclitaxel in the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Yan Diao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - WeiLi Min
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - HuaFeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - ZhiJun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Xijing Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Yang Zhao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China.
| |
Collapse
|
191
|
Pregnane X Receptor Regulates Pathogen-Induced Inflammation and Host Defense against an Intracellular Bacterial Infection through Toll-like Receptor 4. Sci Rep 2016; 6:31936. [PMID: 27550658 PMCID: PMC4994038 DOI: 10.1038/srep31936] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 07/29/2016] [Indexed: 12/20/2022] Open
Abstract
The nuclear pregnane X receptor (PXR) plays a central role in regulating xenobiotic metabolism. We now report a novel role for PXR as a critical negative regulator of innate immunity after infection. Pxr−/− mice exhibited remarkably elevated pro-inflammatory cytokine and chemokine production following infection with Listeria monocytogenes (Lm). Despite the more robust innate immune response, Pxr−/− mice were highly susceptible to Lm infection. Surprisingly, disruption of the Toll-like receptor 4 (TLR4) but not TLR2 signaling restored the inflammation to normal levels and the ability to clear Lm in Pxr−/− mice. Mechanistically, the heightened inflammation in Pxr−/− mice resulted in the death of inflammatory monocytes that led to the enhanced susceptibility to Lm infection. These data demonstrated that PXR regulated pathogen-induced inflammation and host defense against Lm infection through modulating the TLR4 pathway. In summary, we discovered an apical role for PXR in regulating innate immunity. In addition, we uncovered a remarkable negative impact of the TLR4 pathway in controlling the quality of the inflammatory response and host defense against a gram-positive bacterial infection.
Collapse
|
192
|
Ferroptosis is an autophagic cell death process. Cell Res 2016; 26:1021-32. [PMID: 27514700 DOI: 10.1038/cr.2016.95] [Citation(s) in RCA: 1286] [Impact Index Per Article: 142.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/23/2016] [Accepted: 07/25/2016] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated necrosis. It is implicated in various human diseases, including ischemic organ damage and cancer. Here, we report the crucial role of autophagy, particularly autophagic degradation of cellular iron storage proteins (a process known as ferritinophagy), in ferroptosis. Using RNAi screening coupled with subsequent genetic analysis, we identified multiple autophagy-related genes as positive regulators of ferroptosis. Ferroptosis induction led to autophagy activation and consequent degradation of ferritin and ferritinophagy cargo receptor NCOA4. Consistently, inhibition of ferritinophagy by blockage of autophagy or knockdown of NCOA4 abrogated the accumulation of ferroptosis-associated cellular labile iron and reactive oxygen species, as well as eventual ferroptotic cell death. Therefore, ferroptosis is an autophagic cell death process, and NCOA4-mediated ferritinophagy supports ferroptosis by controlling cellular iron homeostasis.
Collapse
|
193
|
Alvarez-Diaz S, Dillon CP, Lalaoui N, Tanzer MC, Rodriguez DA, Lin A, Lebois M, Hakem R, Josefsson EC, O'Reilly LA, Silke J, Alexander WS, Green DR, Strasser A. The Pseudokinase MLKL and the Kinase RIPK3 Have Distinct Roles in Autoimmune Disease Caused by Loss of Death-Receptor-Induced Apoptosis. Immunity 2016; 45:513-526. [PMID: 27523270 DOI: 10.1016/j.immuni.2016.07.016] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 12/12/2022]
Abstract
The kinases RIPK1 and RIPK3 and the pseudo-kinase MLKL have been identified as key regulators of the necroptotic cell death pathway, although a role for MLKL within the whole animal has not yet been established. Here, we have shown that MLKL deficiency rescued the embryonic lethality caused by loss of Caspase-8 or FADD. Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice were viable and fertile but rapidly developed severe lymphadenopathy, systemic autoimmune disease, and thrombocytopenia. These morbidities occurred more rapidly and with increased severity in Casp8(-/-)Mlkl(-/-) and Fadd(-/-)Mlkl(-/-) mice compared to Casp8(-/-)Ripk3(-/-) or Fadd(-/-)Ripk3(-/-) mice, respectively. These results demonstrate that MLKL is an essential effector of aberrant necroptosis in embryos caused by loss of Caspase-8 or FADD. Furthermore, they suggest that RIPK3 and/or MLKL may exert functions independently of necroptosis. It appears that non-necroptotic functions of RIPK3 contribute to the lymphadenopathy, autoimmunity, and excess cytokine production that occur when FADD or Caspase-8-mediated apoptosis is abrogated.
Collapse
Affiliation(s)
- Silvia Alvarez-Diaz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Christopher P Dillon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Najoua Lalaoui
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Maria C Tanzer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Diego A Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ann Lin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Marion Lebois
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Razq Hakem
- Ontario Cancer Institute, University Health Network, and Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| | - Emma C Josefsson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Lorraine A O'Reilly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia.
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia.
| |
Collapse
|
194
|
McLaughlin KA, Nemeth Z, Bradley CA, Humphreys L, Stasik I, Fenning C, Majkut J, Higgins C, Crawford N, Holohan C, Johnston PG, Harrison T, Hanna GG, Butterworth KT, Prise KM, Longley DB. FLIP: A Targetable Mediator of Resistance to Radiation in Non–Small Cell Lung Cancer. Mol Cancer Ther 2016; 15:2432-2441. [DOI: 10.1158/1535-7163.mct-16-0211] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
|
195
|
Nogusa S, Thapa RJ, Dillon CP, Liedmann S, Oguin TH, Ingram JP, Rodriguez DA, Kosoff R, Sharma S, Sturm O, Verbist K, Gough PJ, Bertin J, Hartmann BM, Sealfon SC, Kaiser WJ, Mocarski ES, López CB, Thomas PG, Oberst A, Green DR, Balachandran S. RIPK3 Activates Parallel Pathways of MLKL-Driven Necroptosis and FADD-Mediated Apoptosis to Protect against Influenza A Virus. Cell Host Microbe 2016; 20:13-24. [PMID: 27321907 PMCID: PMC5026823 DOI: 10.1016/j.chom.2016.05.011] [Citation(s) in RCA: 296] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 04/11/2016] [Accepted: 05/16/2016] [Indexed: 12/26/2022]
Abstract
Influenza A virus (IAV) is a lytic virus in primary cultures of many cell types and in vivo. We report that the kinase RIPK3 is essential for IAV-induced lysis of mammalian fibroblasts and lung epithelial cells. Replicating IAV drives assembly of a RIPK3-containing complex that includes the kinase RIPK1, the pseudokinase MLKL, and the adaptor protein FADD, and forms independently of signaling by RNA-sensing innate immune receptors (RLRs, TLRs, PKR), or the cytokines type I interferons and TNF-α. Downstream of RIPK3, IAV activates parallel pathways of MLKL-driven necroptosis and FADD-mediated apoptosis, with the former reliant on RIPK3 kinase activity and neither on RIPK1 activity. Mice deficient in RIPK3 or doubly deficient in MLKL and FADD, but not MLKL alone, are more susceptible to IAV than their wild-type counterparts, revealing an important role for RIPK3-mediated apoptosis in antiviral immunity. Collectively, these results outline RIPK3-activated cytolytic mechanisms essential for controlling respiratory IAV infection.
Collapse
Affiliation(s)
- Shoko Nogusa
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Roshan J Thapa
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Christopher P Dillon
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Swantje Liedmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Thomas H Oguin
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Justin P Ingram
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Diego A Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rachelle Kosoff
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Shalini Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Oliver Sturm
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Katherine Verbist
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - 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
| | - Boris M Hartmann
- Department of Neurology, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA
| | | | | | - Carolina B López
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Andrew Oberst
- Department of Microbiology and Immunology, University of Washington, Seattle, WA 98109, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
196
|
DONG YANRU, BAO CUIFEN, YU JINGWEI, LIU XIA. Receptor-interacting protein kinase 3-mediated programmed cell necrosis in rats subjected to focal cerebral ischemia-reperfusion injury. Mol Med Rep 2016; 14:728-36. [PMID: 27220678 PMCID: PMC4918559 DOI: 10.3892/mmr.2016.5311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 12/22/2015] [Indexed: 01/18/2023] Open
Abstract
In the current study, the activation of tumor necrosis factor-α receptor 1 (TNFR1) and receptor-interacting protein kinase 3 (RIP3) were investigated following cerebral ischemia-reperfusion injury (CIRI). Healthy SD rats were randomly divided into 3 groups: Sham operation group, model group and inhibitor group. The model group and inhibitor group were further divided into 4 subgroups of 6, 12, 24 and 72 h following CIRI. Using right middle cerebral artery embolization, the CIRI model was generated. To confirm that the CIRI model was established, neurological scores, TTC staining and brain water content measurements were conducted. Immunohistochemistry and western blotting were conducted to investigate the expression of TNFR1 and RIP3 in the cerebral cortex. It was observed that nerve cell necrosis occurred following 6 h of CIRI. The appearance of necrotic cells was gradually increased with increasing CIRI duration. TNFR1 and RIP3 were positively expressed following 6 h of CIRI. With increasing durations of CIRI, the protein expression levels of TNFR1 and RIP3 were significantly increased. Pre‑administration with Z-VAD-FMK (zVAD) significantly increased the protein level of RIP3, however, had no effect on the levels of TNFR1, and was accompanied by a reduction in necrosis. In conclusion, RIP3‑mediated cell necrosis was enhanced by caspase blockade zVAD and the function of zVAD was independent of TNFR1 signaling following IR.
Collapse
Affiliation(s)
- YANRU DONG
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - CUIFEN BAO
- Key Laboratory of Molecular Cell Biology and New Drug Development, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - JINGWEI YU
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - XIA LIU
- Department of Histology and Embryology, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
197
|
Al-Lamki RS, Lu W, Manalo P, Wang J, Warren AY, Tolkovsky AM, Pober JS, Bradley JR. Tubular epithelial cells in renal clear cell carcinoma express high RIPK1/3 and show increased susceptibility to TNF receptor 1-induced necroptosis. Cell Death Dis 2016; 7:e2287. [PMID: 27362805 PMCID: PMC5108336 DOI: 10.1038/cddis.2016.184] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 05/26/2016] [Accepted: 05/31/2016] [Indexed: 12/14/2022]
Abstract
We previously reported that renal clear cell carcinoma cells (RCC) express both tumor necrosis factor receptor (TNFR)-1 and -2, but that, in organ culture, a TNF mutein that only engages TNFR1, but not TNFR2, causes extensive cell death. Some RCC died by apoptosis based on detection of cleaved caspase 3 in a minority TUNEL-positive cells but the mechanism of death in the remaining cells was unexplained. Here, we underpin the mechanism of TNFR1-induced cell death in the majority of TUNEL-positive RCC cells, and show that they die by necroptosis. Malignant cells in high-grade tumors displayed threefold to four fold higher expression of both receptor-interacting protein kinase (RIPK)1 and RIPK3 compared with non-tumor kidney tubular epithelium and low-grade tumors, but expression of both enzymes was induced in lower grade tumors in organ culture in response to TNFR1 stimulation. Furthermore, TNFR1 activation induced significant MLKL(Ser358) and Drp1(Ser616) phosphorylation, physical interactions in RCC between RIPK1-RIPK3 and RIPK3-phospho-MLKL(Ser358), and coincidence of phospho-MLKL(ser358) and phospho-Drp1(Ser616) at mitochondria in TUNEL-positive RCC. A caspase inhibitor only partially reduced the extent of cell death following TNFR1 engagement in RCC cells, whereas three inhibitors, each targeting a different step in the necroptotic pathway, were much more protective. Combined inhibition of caspases and necroptosis provided additive protection, implying that different subsets of cells respond differently to TNF-α, the majority dying by necroptosis. We conclude that most high-grade RCC cells express increased amounts of RIPK1 and RIPK3 and are poised to undergo necroptosis in response to TNFR1 signaling.
Collapse
Affiliation(s)
- R S Al-Lamki
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - W Lu
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - P Manalo
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - J Wang
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - A Y Warren
- Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - A M Tolkovsky
- Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - J S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - J R Bradley
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
198
|
Justus SJ, Ting AT. Cloaked in ubiquitin, a killer hides in plain sight: the molecular regulation of RIPK1. Immunol Rev 2016; 266:145-60. [PMID: 26085213 DOI: 10.1111/imr.12304] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the past decade, studies have shown how instrumental programmed cell death (PCD) can be in innate and adaptive immune responses. PCD can be a means to maintain homeostasis, prevent or promote microbial pathogenesis, and drive autoimmune disease and inflammation. The molecular machinery regulating these cell death programs has been examined in detail, although there is still much to be explored. A master regulator of programmed cell death and innate immunity is receptor-interacting protein kinase 1 (RIPK1), which has been implicated in orchestrating various pathologies via the induction of apoptosis, necroptosis, and nuclear factor-κB-driven inflammation. These and other roles for RIPK1 have been reviewed elsewhere. In a reflection of the ability of tumor necrosis factor (TNF) to induce either survival or death response, this molecule in the TNF pathway can transduce either a survival or a death signal. The intrinsic killing capacity of RIPK1 is usually kept in check by the chains of ubiquitin, enabling it to serve in a prosurvival capacity. In this review, the intricate regulatory mechanisms responsible for restraining RIPK1 from killing are discussed primarily in the context of the TNF signaling pathway and how, when these mechanisms are disrupted, RIPK1 is free to unveil its program of cellular demise.
Collapse
Affiliation(s)
- Scott J Justus
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute and Tisch Cancer Institute, New York, NY, USA.,Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adrian T Ting
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute and Tisch Cancer Institute, New York, NY, USA
| |
Collapse
|
199
|
Wang S, Pacher P, De Lisle RC, Huang H, Ding WX. A Mechanistic Review of Cell Death in Alcohol-Induced Liver Injury. Alcohol Clin Exp Res 2016; 40:1215-1223. [PMID: 27130888 PMCID: PMC5455778 DOI: 10.1111/acer.13078] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
Abstract
Alcoholic liver disease (ALD) is a major health problem in the United States and worldwide without successful treatments. Chronic alcohol consumption can lead to ALD, which is characterized by steatosis, inflammation, fibrosis, cirrhosis, and even liver cancer. Recent studies suggest that alcohol induces both cell death and adaptive cell survival pathways in the liver, and the balance of cell death and cell survival ultimately decides the pathogenesis of ALD. This review summarizes the recent progress on the role and mechanisms of apoptosis, necroptosis, and autophagy in the pathogenesis of ALD. Understanding the complex regulation of apoptosis, necrosis, and autophagy may help to develop novel therapeutic strategies by targeting all 3 pathways simultaneously.
Collapse
Affiliation(s)
- Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Robert C. De Lisle
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Heqing Huang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
200
|
Blue Light Action on Mitochondria Leads to Cell Death by Necroptosis. Neurochem Res 2016; 41:2324-35. [DOI: 10.1007/s11064-016-1946-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/28/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
|