251
|
Abstract
Significance: The selenium-containing Glutathione peroxidases (GPxs)1-4 protect against oxidative challenge, inhibit inflammation and oxidant-induced regulated cell death. Recent Advances: GPx1 and GPx4 dampen phosphorylation cascades predominantly via prevention of inactivation of phosphatases by H2O2 or lipid hydroperoxides. GPx2 regulates the balance between regeneration and apoptotic cell shedding in the intestine. It inhibits inflammation-induced carcinogenesis in the gut but promotes growth of established cancers. GPx3 deficiency facilitates platelet aggregation likely via disinhibition of thromboxane biosynthesis. It is also considered a tumor suppressor. GPx4 is expressed in three different forms. The cytosolic form proved to inhibit interleukin-1-driven nuclear factor κB activation and leukotriene biosynthesis. Moreover, it is a key regulator of ferroptosis, because it reduces hydroperoxy groups of complex lipids and silences lipoxygenases. By alternate substrate use, the nuclear form contributes to chromatin compaction. Mitochondrial GPx4 forms the mitochondrial sheath of spermatozoa and, thus, guarantees male fertility. Out of the less characterized GPxs, the cysteine-containing GPx7 and GPx8 are unique in contributing to oxidative protein folding in the endoplasmic reticulum by reacting with protein isomerase as an alternate substrate. A yeast 2-Cysteine glutathione peroxidase equipped with CP and CR was reported to sense H2O2 for inducing an adaptive response. Critical Issues: Most of the findings compiled are derived from tissue culture and/or animal studies only. Their impact on human physiology is sometimes questionable. Future Directions: The expression of individual GPxs and GPx-dependent regulatory phenomena are to be further investigated, in particular in respect to human health.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department of Biochemistry of Micronutrients, German Institute of Human Nutrition-Potsdam-Rehbrücke (DIfE), Nuthetal, Germany
| | - Leopold Flohé
- Depatamento de Biochímica, Universidad de la República, Montevideo, Uruguay.,Dipartimento di Medicina Moleculare, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
252
|
Liu N, Li Y, Chen G, Ge K. Evodiamine induces reactive oxygen species-dependent apoptosis and necroptosis in human melanoma A-375 cells. Oncol Lett 2020; 20:121. [PMID: 32863934 PMCID: PMC7448557 DOI: 10.3892/ol.2020.11983] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
Melanoma is a common solid malignant tumor with a high frequency of metastasis and relapse. Evodiamine (EVO), a natural small molecule, has recently attracted considerable attention due to its pharmacological action, including its anticancer effects. However, the mechanism of the cytotoxic effect exerted by EVO on tumor cells is not yet fully understood. The present study aimed to evaluate the antitumor effects of evodiamine in human melanoma A-375 cells. The results demonstrated that EVO inhibited cell proliferation and induced cell cycle arrest at the G2/M stage in human melanoma A-375 cells. The results also revealed that EVO exposure induced the activation of caspase-3, caspase-9 and poly (ADP-ribose) polymerase 1, as well as mitochondrial membrane potential dissipation in a time-dependent manner, indicating that EVO induced intrinsic apoptosis in A-375 cells. Furthermore, the results revealed that receptor-interacting serine/threonine kinase (RIP) and RIP3 were sequentially activated, suggesting that necroptosis may also be involved in EVO-induced cell death in A-375 cells. In addition, co-treatment with catalase was demonstrated to significantly attenuate the EVO-induced cell death in A-375 cells, indicating that reactive oxygen species (ROS) may serve an important role in EVO-induced cell death. In conclusion, the results of the present study unveiled a novel mechanism of drug action by EVO in human melanoma cells and suggested its potential value in treating human melanoma by inducing cell death via ROS activation.
Collapse
Affiliation(s)
- Ning Liu
- Department of Dermatology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266011, P.R. China.,Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yongxi Li
- Department of Dermatology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266011, P.R. China
| | - Guanzhi Chen
- Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Keli Ge
- Institute of Integrated Medicine, Medical College, Qingdao University, Qingdao, Shandong 266023, P.R. China
| |
Collapse
|
253
|
Liu L, Yan Z, Wang Y, Meng J, Chen G. Suppression of autophagy facilitates hydrogen gas-mediated lung cancer cell apoptosis. Oncol Lett 2020; 20:112. [PMID: 32863925 PMCID: PMC7448556 DOI: 10.3892/ol.2020.11973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/07/2020] [Indexed: 01/10/2023] Open
Abstract
Our previous study found that hydrogen gas (H2) could efficiently inhibit lung cancer progression; however, the underlying mechanisms still remains to be elucidated. The present study aimed to explore the roles of H2 in lung cancer cell autophagy, and reveal the effects of autophagy on H2-mediated lung cancer cell apoptosis and the underlying mechanisms. The expression levels of proteins associated with cell apoptosis and autophagy were detected using western blot analysis. Cell autophagy was inhibited by 3-methyladenine treatment or Beclin1 downregulation, while rapamycin was used to induce autophagy. Cell growth and apoptosis were detected using the Cell Counting Kit-8 and flow cytometry assays, respectively. The results demonstrated that cell apoptosis and autophagy were significantly enhanced in the A549 and H1975 lung cancer cell lines treated with H2. However, autophagy enhancement weakened H2 roles in promoting cell apoptosis and vice versa. In addition, it was found that H2 treatment induced marked decreases in the protein expression levels of phosphorylated STAT3 and Bcl2, and overexpression of STAT3 abolished H2 roles in promoting cell apoptosis and autophagy. Overall, the present study revealed that H2 can promote lung cancer cell apoptosis and autophagy via inhibiting the activation of STAT3/Bcl2 signaling and suppression of autophagy can enhance H2 roles in promoting lung cancer cell apoptosis.
Collapse
Affiliation(s)
- Leyuan Liu
- Department of Respiration, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Zhenfeng Yan
- Department of Respiration, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yuanyuan Wang
- Department of Respiration, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Jinghong Meng
- Department of Respiration, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Gang Chen
- Department of Respiration, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
254
|
The Latest View on the Mechanism of Ferroptosis and Its Research Progress in Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6375938. [PMID: 32908634 PMCID: PMC7474794 DOI: 10.1155/2020/6375938] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/27/2020] [Indexed: 01/08/2023]
Abstract
Ferroptosis is a recently identified nonapoptotic form of cell death whose major markers are iron dependence and accumulation of lipid reactive oxygen species, accompanied by morphological changes such as shrunken mitochondria and increased membrane density. It appears to contribute to the death of tumors, ischemia-reperfusion, acute renal failure, and nervous system diseases, among others. The generative mechanism of ferroptosis includes iron overloading, lipid peroxidation, and downstream execution, while the regulatory mechanism involves the glutathione/glutathione peroxidase 4 pathway, as well as the mevalonate pathway and the transsulfuration pathway. In-depth research has continuously developed and enriched knowledge on the mechanism by which ferroptosis occurs. In recent years, reports of the noninterchangeable role played by selenium in glutathione peroxidase 4 and its function in suppressing ferroptosis and the discovery of ferroptosis suppressor protein 1, identified as a ferroptosis resistance factor parallel to the glutathione peroxidase 4 pathway, have expanded and deepened our understanding of the mechanism by which ferroptosis works. Ferroptosis has been reported in spinal cord injury animal model experiments, and the inhibition of ferroptosis could promote the recovery of neurological function. Here, we review the latest studies on mechanism by which ferroptosis occurs, focusing on the ferroptosis execution and the contents related to selenium and ferroptosis suppressor protein 1. In addition, we summarize the current research status of ferroptosis in spinal cord injury. The aim of this review is to better understand the mechanisms by which ferroptosis occurs and its role in the pathophysiological process of spinal cord injury, so as to provide a new idea and frame of reference for further exploration.
Collapse
|
255
|
Alam S, Abdullah CS, Aishwarya R, Morshed M, Bhuiyan MS. Molecular Perspectives of Mitochondrial Adaptations and Their Role in Cardiac Proteostasis. Front Physiol 2020; 11:1054. [PMID: 32982788 PMCID: PMC7481364 DOI: 10.3389/fphys.2020.01054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the key to properly functioning energy generation in the metabolically demanding cardiomyocytes and thus essential to healthy heart contractility on a beat-to-beat basis. Mitochondria being the central organelle for cellular metabolism and signaling in the heart, its dysfunction leads to cardiovascular disease. The healthy mitochondrial functioning critical to maintaining cardiomyocyte viability and contractility is accomplished by adaptive changes in the dynamics, biogenesis, and degradation of the mitochondria to ensure cellular proteostasis. Recent compelling evidence suggests that the classical protein quality control system in cardiomyocytes is also under constant mitochondrial control, either directly or indirectly. Impairment of cytosolic protein quality control may affect the position of the mitochondria in relation to other organelles, as well as mitochondrial morphology and function, and could also activate mitochondrial proteostasis. Despite a growing interest in the mitochondrial quality control system, very little information is available about the molecular function of mitochondria in cardiac proteostasis. In this review, we bring together current understanding of the adaptations and role of the mitochondria in cardiac proteostasis and describe the adaptive/maladaptive changes observed in the mitochondrial network required to maintain proteomic integrity. We also highlight the key mitochondrial signaling pathways activated in response to proteotoxic stress as a cellular mechanism to protect the heart from proteotoxicity. A deeper understanding of the molecular mechanisms of mitochondrial adaptations and their role in cardiac proteostasis will help to develop future therapeutics to protect the heart from cardiovascular diseases.
Collapse
Affiliation(s)
- Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
256
|
Revisiting Tumors and the Cardiovascular System: Mechanistic Intersections and Divergences in Ferroptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9738143. [PMID: 32908642 PMCID: PMC7450308 DOI: 10.1155/2020/9738143] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/10/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022]
Abstract
Ferroptosis was recently identified as an iron-dependent regulatory necrosis process mediated by polyunsaturated fatty acid (PUFA) peroxidation. The pivotal events related to oxidative stress in ferroptosis include direct or indirect glutathione peroxidase 4 (GPX4) inhibition, ferrous iron overload, and lipid peroxidation. The links between ferroptosis and multiple pathological processes including tumor and cardiovascular system disease have become increasingly apparent, and the mechanisms and compounds involved in ferroptosis, such as reduction of coenzyme Q10 (ubiquinone/CoQ10), are gradually emerging. Current reports have revealed crossroads between ferroptosis and other multiple responses. This overview of the current research illuminates the mechanisms involving ferroptosis-related compounds and emphasizes the crosstalk between ferroptosis and other responses, including mitochondrial damage, endoplasmic reticulum stress (ER stress), autophagy, and the release of damage-associated molecular patterns (DAMPs), to reveal the intersections of regulatory mechanisms. This review also outlines the discovery, characterization, and pathological relevance of ferroptosis and notes controversial elements in ferroptosis-related mechanisms, such as nuclear factor E2-related factor 2 (Nrf2), sequestosome 1 (p62/SQSTM1), and heat shock protein family A member 5 (HSPA5). We hope our inferences will supply a partial reference for disorder prevention and treatment.
Collapse
|
257
|
Sato A, Hiramoto A, Kim HS, Wataya Y. Anticancer Strategy Targeting Cell Death Regulators: Switching the Mechanism of Anticancer Floxuridine-Induced Cell Death from Necrosis to Apoptosis. Int J Mol Sci 2020; 21:ijms21165876. [PMID: 32824286 PMCID: PMC7461588 DOI: 10.3390/ijms21165876] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 01/14/2023] Open
Abstract
Cell death can be broadly characterized as either necrosis or apoptosis, depending on the morphological and biochemical features of the cell itself. We have previously reported that the treatment of mouse mammary carcinoma FM3A cells with the anticancer drug floxuridine (FUdR) induces necrosis in the original clone F28-7 but apoptosis in the variant F28-7-A. We have identified regulators, including heat shock protein 90, lamin-B1, cytokeratin-19, and activating transcription factor 3, of cell death mechanisms by using comprehensive gene and protein expression analyses and a phenotype-screening approach. We also observed that the individual inhibition or knockdown of the identified regulators in F28-7 results in a shift from necrotic to apoptotic morphology. Furthermore, we investigated microRNA (miRNA, miR) expression profiles in sister cell strains F28-7 and F28-7-A using miRNA microarray analyses. We found that several unique miRNAs, miR-351-5p and miR-743a-3p, were expressed at higher levels in F28-7-A than in F28-7. Higher expression of these miRNAs in F28-7 induced by transfecting miR mimics resulted in a switch in the mode of cell death from necrosis to apoptosis. Our findings suggest that the identified cell death regulators may play key roles in the decision of cell death mechanism: necrosis or apoptosis.
Collapse
Affiliation(s)
- Akira Sato
- Department of Biochemistry and Molecular Biology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Correspondence: ; Tel.: +81-4-7121-3620
| | - Akiko Hiramoto
- Division of International Infectious Disease Control, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; (A.H.); (H.-S.K.); (Y.W.)
| | - Hye-Sook Kim
- Division of International Infectious Disease Control, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; (A.H.); (H.-S.K.); (Y.W.)
| | - Yusuke Wataya
- Division of International Infectious Disease Control, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; (A.H.); (H.-S.K.); (Y.W.)
| |
Collapse
|
258
|
Wu RWK, Chu ESM, Yuen JWM, Huang Z. Comparative study of FosPeg® photodynamic effect on nasopharyngeal carcinoma cells in 2D and 3D models. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 210:111987. [PMID: 32801063 DOI: 10.1016/j.jphotobiol.2020.111987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022]
Abstract
Photodynamic Therapy (PDT) offers an alternative option for the treatment of nasopharyngeal carcinoma (NPC). The utilization of 3-dimensional (3D) culture model might provide better understanding of PDT effects on NPC cells. The aim of this in vitro study was to compare PDT effect on NPC cells using 2D and 3D models. Two 3D culture models were established using liquid overlay method with agarose base (MCL) and hanging drop method (MCS). PDT was carried out using the combination of FosPeg® and 652 nm laser in 3D and conventional 2D models. The effects of 3D culture size and morphology on the uptake and distribution of sensitizer and gene expression were examined. Photocytotoxity, mode of cell death, and protein expression were compared for 2D and 3D models. Regular and irregular NPC spheroids were obtained from MCL and MCS methods, respectively. A significantly down-regulation of LMP1 mRNA were observed in MCL spheroid. The sensitizer uptake in 3D spheroids was half of 2D culture. More sensitizers were required to obtain IC50 in 3D spheroids. Apoptosis, necrosis and autophagosomes were detected in PDT treated 2D and 3D cells. Different protein expression patterns were observed in 2D and 3D models. FosPeg® PDT is effective in killing NPC cells. MCL-derived 3D spheroid model is more suitable for the evaluation of PDT killing mechanisms.
Collapse
Affiliation(s)
- Ricky Wing Kei Wu
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China.
| | | | - John Wai Man Yuen
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, China
| | - Zheng Huang
- Biomedical Photonics Center, MOE Key Laboratory of Photonics Science and Technology for Medicine, School of OptoElectronic and Information Engineering, Fujian Normal University, Fuzhou, Hong Kong, China
| |
Collapse
|
259
|
Bougen-Zhukov NM, Lee YY, Lee JYJ, Lee P, Loo LH. PI3K Catalytic Subunits α and β Modulate Cell Death and IL-6 Secretion Induced by Talc Particles in Human Lung Carcinoma Cells. Am J Respir Cell Mol Biol 2020; 62:331-341. [PMID: 31513749 DOI: 10.1165/rcmb.2019-0050oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hydrated magnesium silicate (or "talc" particles) is a sclerosis agent commonly used in the management of malignant pleural effusions, a common symptom of metastatic diseases, including lung cancers. However, the direct effects of talc particles to lung carcinoma cells, which can be found in the malignant pleural effusion fluids from patients with lung cancer, are not fully understood. Here, we report a study of the signaling pathways that can modulate the cell death and IL-6 secretion induced by talc particles in human lung carcinoma cells. We found that talc-sensitive cells have higher mRNA and protein expression of PI3K catalytic subunits α and β. Further experiments confirmed that modulation (inhibition or activation) of the PI3K pathway reduces or enhances cellular sensitivity to talc particles, respectively, independent of the inflammasome. By knocking down specific PI3K isoforms, we also confirmed that both PI3Kα and -β mediate the observed talc effects. Our results suggest a novel role of the PI3K pathway in talc-induced cell death and IL-6 secretion in lung carcinoma cells. These cellular events are known to drive fibrosis, and thus further studies of the PI3K pathway may provide a better understanding of the mechanisms of talc sclerosis in the malignant pleural space.
Collapse
Affiliation(s)
| | - Yin Yeng Lee
- Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Jia-Ying Joey Lee
- Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Pyng Lee
- Division of Respiratory and Critical Care, National University Hospital, National University Health System, Singapore, Singapore; and
| | - Lit-Hsin Loo
- Bioinformatics Institute, Agency for Science, Technology, and Research, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
260
|
Bettadapur A, Ralston KS. Direct and high-throughput assays for human cell killing through trogocytosis by Entamoeba histolytica. Mol Biochem Parasitol 2020; 239:111301. [PMID: 32687867 DOI: 10.1016/j.molbiopara.2020.111301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/08/2023]
Abstract
Entamoeba histolytica is the causative agent of amoebiasis. Pathogenesis is associated with profound damage to human tissues. We previously showed that amoebae kill human cells through trogocytosis. Trogocytosis is likely to underlie tissue damage during infection, although the mechanism is still unknown. Trogocytosis is difficult to assay quantitatively, which makes it difficult to study. Here, we developed two new, complementary assays to measure trogocytosis by quantifying human cell death. One assay uses CellTiterGlo, a luminescent readout for ATP, as a proxy for cell death. We found that the CellTiterGlo could be used to detect death of human cells after co-incubation with amoebae, and that it was sensitive to inhibition of actin or the amoeba surface Gal/GalNAc lectin, two conditions that are known to inhibit amoebic trogocytosis. The other assay uses two fluorescent nuclear stains to directly differentiate live and dead human cells by microscopy, and is also sensitive to inhibition of amoebic trogocytosis through interference with actin. Both assays are simple and inexpensive, can be used with suspension and adherent human cell types, and are amenable to high-throughput approaches. These new assays are tools to improve understanding of trogocytosis and amoebiasis pathogenesis.
Collapse
Affiliation(s)
- Akhila Bettadapur
- Department of Microbiology and Molecular Genetics, University of California, Davis, USA
| | - Katherine S Ralston
- Department of Microbiology and Molecular Genetics, University of California, Davis, USA.
| |
Collapse
|
261
|
dnmt1 function is required to maintain retinal stem cells within the ciliary marginal zone of the zebrafish eye. Sci Rep 2020; 10:11293. [PMID: 32647199 PMCID: PMC7347529 DOI: 10.1038/s41598-020-68016-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/15/2020] [Indexed: 12/22/2022] Open
Abstract
The ciliary marginal zone (CMZ) of the zebrafish retina contains a population of actively proliferating resident stem cells, which generate retinal neurons throughout life. The maintenance methyltransferase, dnmt1, is expressed within the CMZ. Loss of dnmt1 function results in gene misregulation and cell death in a variety of developmental contexts, however, its role in retinal stem cell (RSC) maintenance is currently unknown. Here, we demonstrate that zebrafish dnmt1s872 mutants possess severe defects in RSC maintenance within the CMZ. Using a combination of immunohistochemistry, in situ hybridization, and a transgenic reporter assay, our results demonstrate a requirement for dnmt1 activity in the regulation of RSC proliferation, gene expression and in the repression of endogenous retroelements (REs). Ultimately, cell death is elevated in the dnmt1−/− CMZ, but in a p53-independent manner. Using a transgenic reporter for RE transposition activity, we demonstrate increased transposition in the dnmt1−/− CMZ. Taken together our data identify a critical role for dnmt1 function in RSC maintenance in the vertebrate eye.
Collapse
|
262
|
Tammaro A, Kers J, Scantlebery AML, Florquin S. Metabolic Flexibility and Innate Immunity in Renal Ischemia Reperfusion Injury: The Fine Balance Between Adaptive Repair and Tissue Degeneration. Front Immunol 2020; 11:1346. [PMID: 32733450 PMCID: PMC7358591 DOI: 10.3389/fimmu.2020.01346] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 05/27/2020] [Indexed: 01/10/2023] Open
Abstract
Renal ischemia reperfusion injury (IRI), a common event after renal transplantation, causes acute kidney injury (AKI), increases the risk of delayed graft function (DGF), primes the donor kidney for rejection, and contributes to the long-term risk of graft loss. In the last decade, epidemiological studies have linked even mild episodes of AKI to chronic kidney disease (CKD) progression, and innate immunity seems to play a crucial role. The ischemic insult triggers an acute inflammatory reaction that is elicited by Pattern Recognition Receptors (PRRs), expressed on both infiltrating immune cells as well as tubular epithelial cells (TECs). Among the PRRs, Toll-like receptors (TLRs), their synergistic receptors, Nod-like receptors (NLRs), and the inflammasomes, play a pivotal role in shaping inflammation and TEC repair, in response to renal IRI. These receptors represent promising targets to modulate the extent of inflammation, but also function as gatekeepers of tissue repair, protecting against AKI-to-CKD progression. Despite the important considerations on timely use of therapeutics, in the context of IRI, treatment options are limited by a lack of understanding of the intra- and intercellular mechanisms associated with the activation of innate immune receptors and their impact on adaptive tubular repair. Accumulating evidence suggests that TEC-associated innate immunity shapes the tubular response to stress through the regulation of immunometabolism. Engagement of innate immune receptors provides TECs with the metabolic flexibility necessary for their plasticity during injury and repair. This could significantly affect pathogenic processes within TECs, such as cell death, mitochondrial damage, senescence, and pro-fibrotic cytokine secretion, well-known to exacerbate inflammation and fibrosis. This article provides an overview of the past 5 years of research on the role of innate immunity in experimental and human IRI, with a focus on the cascade of events activated by hypoxic damage in TECs: from programmed cell death (PCD) and mitochondrial dysfunction-mediated metabolic rewiring of TECs to maladaptive repair and progression to fibrosis. Finally, we will discuss the important crosstalk between metabolism and innate immunity observed in TECs and their therapeutic potential in both experimental and clinical research.
Collapse
Affiliation(s)
- Alessandra Tammaro
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Jesper Kers
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.,Biomolecular Systems Analytics, Van 't Hoff Institute for Molecular Sciences (HIMS), University of Amsterdam, Amsterdam, Netherlands
| | - Angelique M L Scantlebery
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam UMC, Amsterdam Infection & Immunity Institute, University of Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
263
|
Uribe P, Meriño J, Manquemilla E, Villagrán C, Vega E, Zambrano F, Schulz M, Pezo F, Villegas JV, Boguen R, Sánchez R. Multiparameter Flow Cytometry Assay for Analysis of Nitrosative Stress Status in Human Spermatozoa. Cytometry A 2020; 97:1238-1247. [DOI: 10.1002/cyto.a.24170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Pamela Uribe
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
- Department of Internal Medicine, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Juan Meriño
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Emilio Manquemilla
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Camila Villagrán
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Etelinda Vega
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Fabiola Zambrano
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Mabel Schulz
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Felipe Pezo
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Juana V. Villegas
- Department of Internal Medicine, Faculty of Medicine Universidad de La Frontera Temuco Chile
- Center of Reproductive Biotechnology ‐ Scientific and Technological Bioresource Nucleus (CEBIOR – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
| | - Rodrigo Boguen
- Departamento de Procesos Diagnósticos y Evaluación, Facultad de Ciencias de la Salud Universidad Católica de Temuco Temuco Chile
| | - Raúl Sánchez
- Center of Excellence in Translational Medicine ‐ Scientific and Technological Bioresource Nucleus (CEMT – BIOREN), Faculty of Medicine Universidad de La Frontera Temuco Chile
- Department of Preclinical Sciences, Faculty of Medicine Universidad de La Frontera Temuco Chile
| |
Collapse
|
264
|
Abstract
Ferroptosis is a recently identified form of regulated cell death that differs from other known forms of cell death morphologically, biochemically, and genetically. The main properties of ferroptosis are free redox-active iron and consequent iron-dependent peroxidation of polyunsaturated fatty acids in cell membrane phospholipids, which results in the accumulation of lipid-based reactive oxygen species due to loss of glutathione peroxidase 4 activity. Ferroptosis has increasingly been associated with neurodegenerative diseases, carcinogenesis, stroke, intracerebral haemorrhage, traumatic brain injury, and ischemia-reperfusion injury. It has also shown a significant therapeutic potential in the treatment of cancer and other diseases. This review summarises current knowledge about and the mechanisms that regulate ferroptosis.
Collapse
|
265
|
Sizemore G, McLaughlin S, Newman M, Brundage K, Ammer A, Martin K, Pugacheva E, Coad J, Mattes MD, Yu HG. Opening large-conductance potassium channels selectively induced cell death of triple-negative breast cancer. BMC Cancer 2020; 20:595. [PMID: 32586284 PMCID: PMC7318490 DOI: 10.1186/s12885-020-07071-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022] Open
Abstract
Background Unlike other breast cancer subtypes that may be treated with a variety of hormonal or targeted therapies, there is a need to identify new, effective targets for triple-negative breast cancer (TNBC). It has recently been recognized that membrane potential is depolarized in breast cancer cells. The primary objective of the study is to explore whether hyperpolarization induced by opening potassium channels may provide a new strategy for treatment of TNBC. Methods Breast cancer datasets in cBioPortal for cancer genomics was used to search for ion channel gene expression. Immunoblots and immunohistochemistry were used for protein expression in culture cells and in the patient tissues. Electrophysiological patch clamp techniques were used to study properties of BK channels in culture cells. Flow cytometry and fluorescence microscope were used for cell viability and cell cycle studies. Ultrasound imaging was used to study xenograft in female NSG mice. Results In large datasets of breast cancer patients, we identified a gene, KCNMA1 (encoding for a voltage- and calcium-dependent large-conductance potassium channel, called BK channel), overexpressed in triple-negative breast cancer patients. Although overexpressed, 99% of channels are closed in TNBC cells. Opening BK channels hyperpolarized membrane potential, which induced cell cycle arrest in G2 phase and apoptosis via caspase-3 activation. In a TNBC cell induced xenograft model, treatment with a BK channel opener significantly slowed tumor growth without cardiac toxicity. Conclusions Our results support the idea that hyperpolarization induced by opening BK channel in TNBC cells can become a new strategy for development of a targeted therapy in TNBC.
Collapse
Affiliation(s)
- Gina Sizemore
- Clinical and Translational Sciences Institute, West Virginia University, Morgantown, USA
| | - Sarah McLaughlin
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Mackenzie Newman
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, 26506, USA
| | - Kathleen Brundage
- Department of Microbiology and Cell Biology, Flow Cytometry Facility, West Virginia University, Morgantown, USA
| | - Amanda Ammer
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Karen Martin
- Animal Models & Imaging Facility, Cancer Institute, West Virginia University, Morgantown, USA
| | - Elena Pugacheva
- Department of Biochemistry, Cancer Institute, West Virginia University, Morgantown, USA
| | - James Coad
- Department of Pathology, West Virginia University, Morgantown, USA
| | - Malcolm D Mattes
- Department of Radiation Oncology, Cancer Institute, West Virginia University, Morgantown, USA
| | - Han-Gang Yu
- Department of Physiology & Pharmacology, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
266
|
Kim J, Chung JY, Park YS, Jang SJ, Kim HR, Choi CM, Song JS. Prognostic Significance of CHIP and RIPK3 in Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:E1496. [PMID: 32521727 PMCID: PMC7352347 DOI: 10.3390/cancers12061496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 11/29/2022] Open
Abstract
RIPK3 is a key regulator of necroptosis, which plays a double-edged sword role in tumor progression. CHIP is an E3 ubiquitin ligase that regulates necroptosis by degrading RIPK3. Here, we investigated the prognostic value of RIPK3 and CHIP expression in 404 patients with non-small cell lung cancer (NSCLC). Expressions of CHIP and RIPK3 showed opposite correlations with survival. CHIP expression was associated with the longer overall survival (OS), whereas RIPK3 expression was associated with the shorter OS. RIPK3 positivity showed marginal association with shorter OS and disease-free survival (DFS) in adjuvant radiotherapy recipients but not in non-recipients, suggesting that necroptosis may induce radioresistance. In multivariate analysis, CHIP expression was associated with longer OS. Compared with other patients, CHIP(-)/RIPK3(+) patients had shorter OS and DFS. In summary, in patients with NSCLC, the expression of CHIP was an independent favorable prognostic factor while that of RIPK3 was an adverse prognostic factor.
Collapse
Affiliation(s)
- Jisup Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.K.); (Y.S.P.); (S.J.J.)
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.K.); (Y.S.P.); (S.J.J.)
| | - Se Jin Jang
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.K.); (Y.S.P.); (S.J.J.)
- Center for Cancer Genome Discovery, Asan Medical Center, Asan Institute for Life Sciences, Seoul 05505, Korea
| | - Hyeong Ryul Kim
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Chang-Min Choi
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan, College of Medicine, Seoul 05505, Korea
- Department of Oncology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul 05505, Korea
| | - Joon Seon Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.K.); (Y.S.P.); (S.J.J.)
| |
Collapse
|
267
|
Mondragon AA, Yalonetskaya A, Ortega AJ, Zhang Y, Naranjo O, Elguero J, Chung WS, McCall K. Lysosomal Machinery Drives Extracellular Acidification to Direct Non-apoptotic Cell Death. Cell Rep 2020; 27:11-19.e3. [PMID: 30943394 PMCID: PMC6613820 DOI: 10.1016/j.celrep.2019.03.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 02/18/2019] [Accepted: 03/08/2019] [Indexed: 02/07/2023] Open
Abstract
Cell death is a fundamental aspect of development, homeostasis, and disease; yet, our understanding of non-apoptotic forms of cell death is limited. One such form is phagoptosis, in which one cell utilizes phagocytosis machinery to kill another cell that would otherwise continue living. We have previously identified a non-autonomous requirement of phagocytosis machinery for the developmental programmed cell death of germline nurse cells in the Drosophila ovary; however, the precise mechanism of death remained elusive. Here, we show that lysosomal machinery acting in epithelial follicle cells is used to non-autonomously induce the death of nearby germline cells. Stretch follicle cells recruit V-ATPases and chloride channels to their plasma membrane to extracellularly acidify the germline and release cathepsins that destroy the nurse cells. Our results reveal a role for lysosomal machinery acting at the plasma membrane to cause the death of neighboring cells, providing insight into mechanisms driving non-autonomous cell death. Mondragon et al. show that V-ATPase proton pumps localize to the plasma membrane of follicle cells and promote extracellular acidification to eliminate adjacent nurse cells in the Drosophila ovary. The follicle cells subsequently release cathepsins by exocytosis into the nurse cells to promote their final degradation.
Collapse
Affiliation(s)
- Albert A Mondragon
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA 02215, USA
| | - Alla Yalonetskaya
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Anthony J Ortega
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Yuanhang Zhang
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Oandy Naranjo
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Johnny Elguero
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Won-Suk Chung
- Department of Biological Sciences, KAIST, Daejeon, South Korea
| | - Kimberly McCall
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA.
| |
Collapse
|
268
|
Marchetti P, Suleiman M, De Luca C, Baronti W, Bosi E, Tesi M, Marselli L. A direct look at the dysfunction and pathology of the β cells in human type 2 diabetes. Semin Cell Dev Biol 2020; 103:83-93. [PMID: 32417220 DOI: 10.1016/j.semcdb.2020.04.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/27/2020] [Accepted: 04/09/2020] [Indexed: 12/25/2022]
Abstract
β cells uniquely produce and secrete insulin under the control of several, integrated signals, to maintain blood glucose concentrations within a narrow physiological interval. β cell failure is key to the onset and progression of type 2 diabetes, due to impaired function and reduced mass. In this review we focus on several features of human β cell dysfunction and pathology in type 2 diabetes, as revealed by direct assessment of isolated islet traits and examination of pancreatic tissue from organ donors, surgical samples or autoptic specimens. Insulin secretion defects and pathology findings are discussed in relation to some of the major underlying mechanisms, to also provide clues for conceiving better prevention and treatment of type 2 diabetes by targeting the pancreatic β cells.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy.
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| | - Walter Baronti
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| | - Emanuele Bosi
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| | - Marta Tesi
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine - University of Pisa, Via Savi 10, Pisa, Italy
| |
Collapse
|
269
|
Correcting an instance of synthetic lethality with a pro-survival sequence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118734. [PMID: 32389645 DOI: 10.1016/j.bbamcr.2020.118734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/11/2020] [Accepted: 05/02/2020] [Indexed: 11/21/2022]
Abstract
A human cDNA encoding the LIM domain containing 194 amino acid cysteine and glycine rich protein 3 (CSRP3) was identified as a BAX suppressor in yeast and a pro-survival sequence that abrogated copper mediated regulated cell death (RCD). Yeast lacks a CSRP3 orthologue but it has four LIM sequences, namely RGA1, RGA2, LRG1 and PXL1. These are known regulators of stress responses yet their roles in RCD remain unknown. Given that LIMs interact with other LIMs, we ruled out the possibility that overexpressed yeast LIMs alone could prevent RCD and that CSRP3 functions by acting as a dominant regulator of yeast LIMs. Of interest was the discovery that even though yeast cells lacking the LIM encoding PXL1 had no overt growth defect, it was nevertheless supersensitive to the effects of sublethal levels of copper. Heterologous expression of human CSPR3 as well as the pro-survival 14-3-3 sequence corrected this copper supersensitivity. These results show that the pxl1∆-copper synthetic lethality is likely due to the induction of RCD. This differs from the prevailing model in which synthetic lethality occurs because of specific defects generated by the combined loss of two overlapping but non-essential functions.
Collapse
|
270
|
The mystery of mitochondria-ER contact sites in physiology and pathology: A cancer perspective. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165834. [PMID: 32437958 DOI: 10.1016/j.bbadis.2020.165834] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria-associated membranes (MAM), physical platforms that enable communication between mitochondria and the endoplasmic reticulum (ER), are enriched with many proteins and enzymes involved in several crucial cellular processes, such as calcium (Ca2+) homeostasis, lipid synthesis and trafficking, autophagy and reactive oxygen species (ROS) production. Accumulating studies indicate that tumor suppressors and oncogenes are present at these intimate contacts between mitochondria and the ER, where they influence Ca2+ flux between mitochondria and the ER or affect lipid homeostasis at MAM, consequently impacting cell metabolism and cell fate. Understanding these fundamental roles of mitochondria-ER contact sites as important domains for tumor suppressors and oncogenes can support the search for new and more precise anticancer therapies. In the present review, we summarize the current understanding of basic MAM biology, composition and function and discuss the possible role of MAM-resident oncogenes and tumor suppressors.
Collapse
|
271
|
McNeill SM, Giles NM, Preston D, Jones PP, Crowley JD, Giles GI. Quadruply Stranded Metallo-Supramolecular Helicate [Pd 2(hextrz) 4] 4+ Acts as a Molecular Mimic of Cytolytic Peptides. Chem Res Toxicol 2020; 33:1822-1834. [PMID: 32347099 DOI: 10.1021/acs.chemrestox.0c00061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Pd2(hextrz)4]4+ is a quadruply stranded helicate, a novel bioinorganic complex designed to mimic the structure and function of proteins due to its high stability and supramolecular size. We have previously reported that [Pd2(hextrz)4]4+ exhibited cytotoxicity toward a range of cell lines, with IC50 values ranging from 3 to 10 μM. Here we demonstrate that [Pd2(hextrz)4]4+ kills cells by forming pores within the cell membrane, a mechanism of cell death analogous to the naturally occurring cytolytic peptides. [Pd2(hextrz)4]4+ induced cell death is characterized by an initial influx of Ca2+, followed by nuclear condensation and mitochondrial swelling. This is accompanied by progressive cell membrane damage that results in the formation of large blebs at the cell surface. This allows the efflux of molecules from the cell leading to loss of cell viability. These data suggest that it may be possible to design metallo-supramolecular complexes to mimic the cytotoxic action of pore forming proteins and peptides and so provide a new class of drug to treat cancer, autoimmune disorders, and microbial infection.
Collapse
Affiliation(s)
- Samantha M McNeill
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Niroshini M Giles
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Dan Preston
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Peter P Jones
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - James D Crowley
- Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Gregory I Giles
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
272
|
Abstract
Currently the study of Regulated Cell Death (RCD) processes is limited to the use of lysed cell populations for Western blot analysis of each separate RCD process. We have previously shown that intracellular antigen flow cytometric analysis of RIP3, Caspase-3 and cell viability dye allowed the determination of levels of apoptosis (Caspase-3+ ve/RIP3− ve), necroptosis (RIP3Hi + ve/Caspase-3− ve) and RIP1-dependent apoptosis (Caspase-3+ ve/RIP3+ ve) in a single Jurkat cell population. The addition of more intracellular markers allows the determination of the incidence of parthanatos (PARP), DNA Damage Response (DDR, H2AX), H2AX hyper-activation of PARP (H2AX/PARP) autophagy (LC3B) and ER stress (PERK), thus allowing the identification of 124 sub-populations both within live and dead cell populations. Shikonin simultaneously induced Jurkat cell apoptosis and necroptosis the degree of which can be shown flow cytometrically together with the effects of blockade of these forms of cell death by zVAD and necrostatin-1 have on specific RCD populations including necroptosis, early and late apoptosis and RIP1-dependent apoptosis phenotypes in live and dead cells. Necrostatin-1 and zVAD was shown to modulate levels of shikonin induced DDR, hyper-action of PARP and parthanatos in the four forms of RCD processes analysed. LC3B was up-regulated by combined treatment of zVAD with chloroquine which also revealed that DNA damage was reduced in live cells but enhanced in dead cells indicating the role of autophagy in maintaining cell health. This approach to RCD research should be a great advance to understanding the mechanisms of drugs and their effects upon RCD populations.
Collapse
|
273
|
Madorran E, Stožer A, Bevc S, Maver U. In vitro toxicity model: Upgrades to bridge the gap between preclinical and clinical research. Bosn J Basic Med Sci 2020; 20:157-168. [PMID: 31621554 PMCID: PMC7202182 DOI: 10.17305/bjbms.2019.4378] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/29/2019] [Indexed: 11/30/2022] Open
Abstract
The Centers for Disease Control and Prevention (CDC) provides extensive data that indicate our need for drugs to maintain human population health. Despite the substantial availability of drugs on the market, many patients lack specific drugs. New drugs are required to tackle this issue. Moreover, we need more reliable models for testing drug toxicity, as too many drug approval failures occur with the current models. This article briefly describes various approaches of the currently used models for toxicity screening, to justify the selection of in vitro cell-based models. Cell-based toxicity models have the best potential to reliably predict drug toxicity in humans, as they are developed using the cells of the target organism. However, currently, a large gap exists between in vitro cell-based approach to toxicity testing and the clinical approach, which may be contributing to drug approval failures. We propose improvements to in vitro cell-based toxicity models, which is often an insight approach, to better match this approach with the clinical homeostatic approach. This should enable a more accurate comparison of data between the preclinical as well as clinical models and provide a more comprehensive understanding of human physiology and biological effects of drugs.
Collapse
Affiliation(s)
- Eneko Madorran
- Institute of Anatomy, Histology and Embryology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Sebastjan Bevc
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Nephrology, Clinic for Internal Medicine, University Medical Center Maribor, Maribor, Slovenia
| | - Uroš Maver
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
274
|
Preface: Life through death-Key role of cellular suicide for colonial and organismal homeostasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:xi-xv. [PMID: 32334819 DOI: 10.1016/s1937-6448(20)30047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
|
275
|
A drug potency signature links progression of chronic lymphocytic leukemia to mitochondria-related stress responses and metabolic reprogramming under hypoxia. Toxicol Appl Pharmacol 2020; 398:115016. [PMID: 32325063 DOI: 10.1016/j.taap.2020.115016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 11/23/2022]
Abstract
Chronic lymphocytic leukemia (CLL) cells change their metabolic program between normoxia and hypoxia, possibly affecting cytotoxic drug potency by altering mitochondria-related cell stress responses (MRCSR) including mitophagy, mitochondrial biogenesis, and mitochondrial proteostasis. We evaluated in CLL cells from nine patients, the single and multiple-combined drug potency of arsenic trioxide (ATO), valproic acid (VPA), vincristine (VCR) and MG132 as four pharmacological sensors influencing mitochondrial apoptosis, mitochondrial biogenesis, mitophagy, and mitochondrial proteostasis respectively, under normoxia and hypoxia to force hypoxia-induced metabolic reprogramming (HMR). Untreated cells from all patients remained viable under O2 levels below 0.5% for 72 h. We obtained 21 measures of drug potency and interaction at 50% effect level that we denoted drug potency signature (DPS). Using the comparative DPS between normoxia and hypoxia, two non-supervised classification algorithms discriminated CLL patients with active disease (ADT) and stable disease (NAD) and showed complete consistency with their clinical characteristics. In ADT group under hypoxia, the potency of MG132 was increased, the interaction of ATO + VPA and ATO + VPA + VCR shifted towards antagonism, and ATO + VPA + VCR + MG132 shifted towards synergism, indicating a prominent role of mitochondrial proteostasis. Classification of patients based on DPS, depended on the contrasting response of drugs under hypoxia and normoxia, owing to HMR. Using these drugs as pharmacological sensors, we linked the metabolic arrangement of CLL cells under hypoxia, to potency of drugs targeting MRCSR, and to the clinical features of individual patients, therefore providing new sources of data on disease progression, drug response and risk prognosis.
Collapse
|
276
|
Sarcognato S, de Jong IEM, Fabris L, Cadamuro M, Guido M. Necroptosis in Cholangiocarcinoma. Cells 2020; 9:cells9040982. [PMID: 32326539 PMCID: PMC7226990 DOI: 10.3390/cells9040982] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
Necroptosis is a type of regulated cell death that is increasingly being recognized as a relevant pathway in different pathological conditions. Necroptosis can occur in response to multiple stimuli, is triggered by the activation of death receptors, and is regulated by receptor-interacting protein kinases 1 and 3 and mixed-lineage kinase domain-like, which form a regulatory complex called the necrosome. Accumulating evidence suggests that necroptosis plays a complex role in cancer, which is likely context-dependent and can vary among different types of neoplasms. Necroptosis serves as an alternative mode of programmed cell death overcoming apoptosis and, as a pro-inflammatory death type, it may inhibit tumor progression by releasing damage-associated molecular patterns to elicit robust cross-priming of anti-tumor CD8+ T cells. The development of therapeutic strategies triggering necroptosis shows great potential for anti-cancer therapy. In this review, we summarize the current knowledge on necroptosis and its role in liver biliary neoplasms, underlying the potential of targeting necroptosis components for cancer treatment.
Collapse
Affiliation(s)
- Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - Iris E. M. de Jong
- Department of Surgery, Section of Hepatobiliary Surgery and Liver Transplantation, University Medical Center Groningen, 9700 Groningen, The Netherlands
| | - Luca Fabris
- Department of Molecular Medicine—DMM, University of Padova, 35121 Padova, Italy
| | | | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
- Department of Medicine—DIMED, University of Padova, 35121 Padova, Italy
- Correspondence: ; Tel.: +39-0422-322750
| |
Collapse
|
277
|
Doll NM, Just J, Brunaud V, Caïus J, Grimault A, Depège-Fargeix N, Esteban E, Pasha A, Provart NJ, Ingram GC, Rogowsky PM, Widiez T. Transcriptomics at Maize Embryo/Endosperm Interfaces Identifies a Transcriptionally Distinct Endosperm Subdomain Adjacent to the Embryo Scutellum. THE PLANT CELL 2020; 32:833-852. [PMID: 32086366 PMCID: PMC7145466 DOI: 10.1105/tpc.19.00756] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/03/2020] [Accepted: 02/20/2020] [Indexed: 05/23/2023]
Abstract
Seeds are complex biological systems comprising three genetically distinct tissues nested one inside another (embryo, endosperm, and maternal tissues). However, the complexity of the kernel makes it difficult to understand intercompartment interactions without access to spatially accurate information. Here, we took advantage of the large size of the maize (Zea mays) kernel to characterize genome-wide expression profiles of tissues at different embryo/endosperm interfaces. Our analysis identifies specific transcriptomic signatures in two interface tissues compared with whole seed compartments: the scutellar aleurone layer and the newly named endosperm adjacent to scutellum (EAS). The EAS, which appears around 9 d after pollination and persists for around 11 d, is confined to one to three endosperm cell layers adjacent to the embryonic scutellum. Its transcriptome is enriched in genes encoding transporters. The absence of the embryo in an embryo specific mutant can alter the expression pattern of EAS marker genes. The detection of cell death in some EAS cells together with an accumulation of crushed cell walls suggests that the EAS is a dynamic zone from which cell layers in contact with the embryo are regularly eliminated and to which additional endosperm cells are recruited as the embryo grows.
Collapse
Affiliation(s)
- Nicolas M Doll
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Jeremy Just
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Véronique Brunaud
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, F-91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, F-91405 Orsay, France
| | - José Caïus
- Institute of Plant Sciences Paris-Saclay IPS2, CNRS, INRA, Université Paris-Sud, Université Evry, Université Paris-Saclay, F-91405 Orsay, France
- Institute of Plant Sciences Paris-Saclay IPS2, Paris Diderot, Sorbonne Paris-Cité, F-91405 Orsay, France
| | - Aurélie Grimault
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Nathalie Depège-Fargeix
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Eddi Esteban
- Department of Cell and Systems Biology/Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario M5S 3B2, Canada
| | - Asher Pasha
- Department of Cell and Systems Biology/Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario M5S 3B2, Canada
| | - Nicholas J Provart
- Department of Cell and Systems Biology/Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, Ontario M5S 3B2, Canada
| | - Gwyneth C Ingram
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Peter M Rogowsky
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| | - Thomas Widiez
- Laboratoire Reproduction et Développement des Plantes, Univ Lyon, ENS de Lyon, UCB Lyon 1, CNRS, INRAE, F-69342 Lyon, France
| |
Collapse
|
278
|
Life, death, and autophagy in cancer: NF-κB turns up everywhere. Cell Death Dis 2020; 11:210. [PMID: 32231206 PMCID: PMC7105474 DOI: 10.1038/s41419-020-2399-y] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022]
Abstract
Escaping programmed cell death is a hallmark of cancer. NF-κB transcription factors are key regulator of cell survival and aberrant NF-κB signaling has been involved in the pathogenesis of most human malignancies. Although NF-κB is best known for its antiapoptotic role, other processes regulating the life/death balance, such as autophagy and necroptosis, seem to network with NF-κB. This review discusses how the reciprocal regulation of NF-κB, autophagy and programmed cell death affect cancer development and progression.
Collapse
|
279
|
Han C, Liu Y, Dai R, Ismail N, Su W, Li B. Ferroptosis and Its Potential Role in Human Diseases. Front Pharmacol 2020; 11:239. [PMID: 32256352 PMCID: PMC7090218 DOI: 10.3389/fphar.2020.00239] [Citation(s) in RCA: 197] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 12/26/2022] Open
Abstract
Ferroptosis is a novel regulated cell death pattern discovered when studying the mechanism of erastin-killing RAS mutant tumor cells in 2012. It is an iron-dependent programmed cell death pathway mainly caused by an increased redox imbalance but with distinct biological and morphology characteristics when compared to other known cell death patterns. Ferroptosis is associated with various diseases including acute kidney injury, cancer, and cardiovascular, neurodegenerative, and hepatic diseases. Moreover, activation or inhibition of ferroptosis using a variety of ferroptosis initiators and inhibitors can modulate disease progression in animal models. In this review, we provide a comprehensive analysis of the characteristics of ferroptosis, its initiators and inhibitors, and the potential role of its main metabolic pathways in the treatment and prevention of various diseased states. We end the review with the current knowledge gaps in this area to provide direction for future research on ferroptosis.
Collapse
Affiliation(s)
- Chu Han
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Yuanyuan Liu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Nafissa Ismail
- Neuroimmunology, Stress and Endocrinology (NISE) Lab, School of Psychology, Faculty of Social Science, University of Ottawa, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin, China
| | - Bo Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
280
|
Ruan J, Wang S, Wang J. Mechanism and regulation of pyroptosis-mediated in cancer cell death. Chem Biol Interact 2020; 323:109052. [PMID: 32169591 DOI: 10.1016/j.cbi.2020.109052] [Citation(s) in RCA: 169] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/29/2020] [Accepted: 03/09/2020] [Indexed: 12/18/2022]
Abstract
Pyroptosis, a form of programmed cell death, has garnered increasing attention as it relates to innate immunity and diseases. The discovery of caspase-1/3/4/5/8/11 function in sensing various challenges expands the spectrum of pyroptosis mediators and also reveals that pyroptosis is not cell type specific. Recent studies have identified that pyroptosis has become a new topic in cancer research because it may affect all stages of carcinogenesis. In this mini-review, we provided a primer on pyroptosis, discussed the induction of pyroptosis in cancer and its implications in cancer management. Moreover, its two important executioners, the gasdermin D (GSDMD) and gasdermin E (GSDME), the functions and mechanisms of them involved in the regulation of cancer therapy were focused on. Small molecules-mediated pyroptosis were found to effectively inhibit various tumor cells. In brief, the findings of pyroptosis-dependent cancer progression, new drugs and therapeutic targets may lead to a promising, novel therapeutic approach for cancer patients.
Collapse
Affiliation(s)
- Jianwei Ruan
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, 318000, China.
| | - Shijian Wang
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, 318000, China
| | - Jiabing Wang
- Municipal Hospital Affiliated to Medical School of Taizhou University, Taizhou, 318000, China.
| |
Collapse
|
281
|
Galluzzi L, Vitale I, Warren S, Adjemian S, Agostinis P, Martinez AB, Chan TA, Coukos G, Demaria S, Deutsch E, Draganov D, Edelson RL, Formenti SC, Fucikova J, Gabriele L, Gaipl US, Gameiro SR, Garg AD, Golden E, Han J, Harrington KJ, Hemminki A, Hodge JW, Hossain DMS, Illidge T, Karin M, Kaufman HL, Kepp O, Kroemer G, Lasarte JJ, Loi S, Lotze MT, Manic G, Merghoub T, Melcher AA, Mossman KL, Prosper F, Rekdal Ø, Rescigno M, Riganti C, Sistigu A, Smyth MJ, Spisek R, Stagg J, Strauss BE, Tang D, Tatsuno K, van Gool SW, Vandenabeele P, Yamazaki T, Zamarin D, Zitvogel L, Cesano A, Marincola FM. Consensus guidelines for the definition, detection and interpretation of immunogenic cell death. J Immunother Cancer 2020; 8:e000337. [PMID: 32209603 PMCID: PMC7064135 DOI: 10.1136/jitc-2019-000337] [Citation(s) in RCA: 670] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York City, New York, USA
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Université de Paris, Paris, France
| | - Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Sarah Warren
- NanoString Technologies, Seattle, Washington, USA
| | - Sandy Adjemian
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB-KU Leuven Center for Cancer Biology, KU Leuevn, Leuven, Belgium
| | - Aitziber Buqué Martinez
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - George Coukos
- Ludwig Institute for Cancer Research and Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, New York, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM "Molecular Radiotherapy and therapeutic innovation", U1030 Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- SIRIC SOCRATES, DHU Torino, Faculté de Medecine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Richard L Edelson
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jitka Fucikova
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - Lucia Gabriele
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Udo S Gaipl
- Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Encouse Golden
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
- Sandra and Edward Meyer Cancer Center, New York City, New York, USA
| | - Jian Han
- iRepertoire, Inc, Huntsville, Alabama, USA
| | - Kevin J Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital/Institute of Cancer Research National Institute for Health Biomedical Research Centre, London, UK
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, National Cancer Institute/Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Tim Illidge
- University of Manchester, NIHR Manchester Biomedical Research Centre, Christie Hospital, Manchester, UK
| | - Michael Karin
- Department of Pharmacology and Pathology, University of California at San Diego (UCSD) School of Medicine, La Jolla, California, USA
| | - Howard L Kaufman
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Replimune, Inc, Woburn, Massachusetts, USA
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Guido Kroemer
- Université de Paris, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1138, Paris, France
- Sorbonne Université, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
| | - Juan Jose Lasarte
- Program of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA), University of Navarra, Pamplona, Spain
| | - Sherene Loi
- Division of Research and Clinical Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gwenola Manic
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS, Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, MSKCC, New York City, New York, USA
- Weill Cornell Medical College, New York City, New York, USA
- Parker Institute for Cancer Immunotherapy, MSKCC, New York City, New York, USA
| | | | | | - Felipe Prosper
- Hematology and Cell Therapy, Clinica Universidad de Navarra, Pamplona, Spain
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
- Interdepartmental Research Center of Molecular Biotechnology, University of Torino, Torino, Italy
| | - Antonella Sistigu
- UOSD Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Istituto di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Radek Spisek
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
- Sotio, Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec City, Canada
- Institut du Cancer de Montréal, Montréal, Quebec City, Canada
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Quebec City, Canada
| | - Bryan E Strauss
- Centro de Investigação Translacional em Oncologia/LIM24, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brasil
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Kazuki Tatsuno
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
- Methusalem program, Ghent University, Ghent, Belgium
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York City, New York, USA
| | - Dmitriy Zamarin
- Department of Medicine, Weill Cornell Medical College, New York City, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Equipe labellisée par la Ligue contre le cancer, Gustave Roussy, Villejuif, France
- Faculty of Medicine, University of Paris Sud/Paris Saclay, Le Kremlin-Bicêtre, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | | | | |
Collapse
|
282
|
Cellular Interplay as a Consequence of Inflammatory Signals Leading to Liver Fibrosis Development. Cells 2020; 9:cells9020461. [PMID: 32085494 PMCID: PMC7072785 DOI: 10.3390/cells9020461] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/10/2020] [Accepted: 02/15/2020] [Indexed: 02/06/2023] Open
Abstract
Inflammation has been known to be an important driver of fibrogenesis in the liver and onset of hepatic fibrosis. It starts off as a process meant to protect the liver from further damage, but it can become the main promoter of liver fibrosis. There are many inflammation-related pathways activated during liver fibrosis that lead to hepatic stellate cells (HSCs) activation and collagen-deposition in the liver. Such events are mostly modulated upstream of HSCs and involve signals from hepatocytes and innate immune cells. One particular event is represented by cell death during liver injury that generates multiple inflammatory signals that further trigger sterile inflammation and enhancement of inflammatory response. The assembly of inflammasome that responds to danger-associated molecular patterns (DAMPs) stimulates the release of pro-inflammatory cytokines and at the same time, initiates programmed cell death called pyroptosis. This review focuses on cellular and molecular mechanisms responsible for initiation and progress of inflammation in the liver.
Collapse
|
283
|
Melittin-A Natural Peptide from Bee Venom Which Induces Apoptosis in Human Leukaemia Cells. Biomolecules 2020; 10:biom10020247. [PMID: 32041197 PMCID: PMC7072249 DOI: 10.3390/biom10020247] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/29/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Bee venom is a very complex mixture produced and secreted by the honeybee (Apis mellifera). Melittin is a major component of bee venom that accounts for about 52% of its dry mass. A vast number of studies have been dedicated to the effects of melittin’s regulation of apoptosis and to the factors that induce apoptosis in various types of cancer such as breast, ovarian, prostate, lung. The latest evidence indicates its potential as a therapeutic agent in the treatment of leukaemia. The aim of our present study is to evaluate melittin’s ability to induce apoptosis in leukaemia cell lines of different origin acute lymphoblastic leukaemia (CCRF-CEM) and chronic myelogenous leukaemia (K-562). We demonstrated that melittin strongly reduced cell viability in both leukaemia cell lines but not in physiological peripheral blood mononuclear cells (PMBCs). Subsequent estimated parameters (mitochondrial membrane potential, Annexin V binding and Caspases 3/7 activity) clearly demonstrated that melittin induced apoptosis in leukaemia cells. This is a very important step for research into the development of new potential anti-leukaemia as well as anticancer therapies. Further analyses on the molecular level have been also planned (analysis of proapoptotic genes expression and DNA damages) for our next research project, which will also focus on melittin.
Collapse
|
284
|
Rodriguez-Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L. Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol 2020; 21:120-134. [PMID: 31873291 DOI: 10.1038/s41590-019-0561-4] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022]
Abstract
Therapeutic irradiation of the tumor microenvironment causes differential activation of pro-survival and pro-death pathways in malignant, stromal, endothelial and immune cells, hence causing a profound cellular and biological reconfiguration via multiple, non-redundant mechanisms. Such mechanisms include the selective elimination of particularly radiosensitive cell types and consequent loss of specific cellular functions, the local release of cytokines and danger signals by dying radiosensitive cells, and altered cytokine secretion by surviving radioresistant cells. Altogether, these processes create chemotactic and immunomodulatory cues for incoming and resident immune cells. Here we discuss how cytoprotective and cytotoxic signaling modules activated by radiation in specific cell populations reshape the immunological tumor microenvironment.
Collapse
Affiliation(s)
- Maria Esperanza Rodriguez-Ruiz
- Department of Radiation Oncology, University of Navarra Clinic, Pamplona, Spain
- Centro de Investigación Médica Aplicada (CIMA), Pamplona, Spain
| | - Ilio Vitale
- IIGM-Italian Institute for Genomic Medicine, c/o IRCCS Candiolo, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Kevin J Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital/Institute of Cancer Research National Institute for Health Biomedical Research Centre, London, UK
| | - Ignacio Melero
- Department of Radiation Oncology, University of Navarra Clinic, Pamplona, Spain
- Centro de Investigación Médica Aplicada (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
- Université de Paris, Paris, France.
| |
Collapse
|
285
|
Lotsberg ML, Wnuk-Lipinska K, Terry S, Tan TZ, Lu N, Trachsel-Moncho L, Røsland GV, Siraji MI, Hellesøy M, Rayford A, Jacobsen K, Ditzel HJ, Vintermyr OK, Bivona TG, Minna J, Brekken RA, Baguley B, Micklem D, Akslen LA, Gausdal G, Simonsen A, Thiery JP, Chouaib S, Lorens JB, Engelsen AST. AXL Targeting Abrogates Autophagic Flux and Induces Immunogenic Cell Death in Drug-Resistant Cancer Cells. J Thorac Oncol 2020; 15:973-999. [PMID: 32018052 PMCID: PMC7397559 DOI: 10.1016/j.jtho.2020.01.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/29/2019] [Accepted: 01/19/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Acquired cancer therapy resistance evolves under selection pressure of immune surveillance and favors mechanisms that promote drug resistance through cell survival and immune evasion. AXL receptor tyrosine kinase is a mediator of cancer cell phenotypic plasticity and suppression of tumor immunity, and AXL expression is associated with drug resistance and diminished long-term survival in a wide range of malignancies, including NSCLC. METHODS We aimed to investigate the mechanisms underlying AXL-mediated acquired resistance to first- and third-generation small molecule EGFR tyrosine kinase inhibitors (EGFRi) in NSCLC. RESULTS We found that EGFRi resistance was mediated by up-regulation of AXL, and targeting AXL reduced reactivation of the MAPK pathway and blocked onset of acquired resistance to long-term EGFRi treatment in vivo. AXL-expressing EGFRi-resistant cells revealed phenotypic and cell signaling heterogeneity incompatible with a simple bypass signaling mechanism, and were characterized by an increased autophagic flux. AXL kinase inhibition by the small molecule inhibitor bemcentinib or siRNA mediated AXL gene silencing was reported to inhibit the autophagic flux in vitro, bemcentinib treatment blocked clonogenicity and induced immunogenic cell death in drug-resistant NSCLC in vitro, and abrogated the transcription of autophagy-associated genes in vivo. Furthermore, we found a positive correlation between AXL expression and autophagy-associated gene signatures in a large cohort of human NSCLC (n = 1018). CONCLUSION Our results indicate that AXL signaling supports a drug-resistant persister cell phenotype through a novel autophagy-dependent mechanism and reveals a unique immunogenic effect of AXL inhibition on drug-resistant NSCLC cells.
Collapse
Affiliation(s)
- Maria L Lotsberg
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Katarzyna Wnuk-Lipinska
- Department of Biomedicine, University of Bergen, Bergen, Norway; BerGenBio ASA, Bergen, Norway
| | - Stéphane Terry
- INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ning Lu
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Laura Trachsel-Moncho
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gro V Røsland
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | | | | - Austin Rayford
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kirstine Jacobsen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Henrik J Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Olav K Vintermyr
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Trever G Bivona
- Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - John Minna
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Surgery, Pharmacology and Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Surgery, Pharmacology and Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Bruce Baguley
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jean Paul Thiery
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Biomedical Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, A-STAR, Singapore; Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China; Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong
| | - Salem Chouaib
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Thumbay Research Institute for Precision Medicine, GMU Ajman, United Arab Emirates
| | - James B Lorens
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Agnete Svendsen Tenfjord Engelsen
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway; INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
286
|
Lee SH, Kwon JY, Moon J, Choi J, Jhun J, Jung K, Cho KH, Darlami O, Lee HH, Jung ES, Shin DY, Lee BI, Cho ML. Inhibition of RIPK3 Pathway Attenuates Intestinal Inflammation and Cell Death of Inflammatory Bowel Disease and Suppresses Necroptosis in Peripheral Mononuclear Cells of Ulcerative Colitis Patients. Immune Netw 2020; 20:e16. [PMID: 32395368 PMCID: PMC7192830 DOI: 10.4110/in.2020.20.e16] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/03/2020] [Accepted: 01/09/2020] [Indexed: 01/11/2023] Open
Abstract
Receptor-interacting serine/threonine-protein kinase (RIPK) 3 is a member of the TNF receptor-I signaling complex and mediates necroptosis, an inflammatory cell death. Ulcerative colitis (UC) is an excessive inflammatory disease caused by uncontrolled T cell activation. The current study is aimed to determine whether RIPK3 inhibitor attenuates UC development inhibiting inflammation and necroptosis using experimental colitis mice model. Dextran sulfate sodium-induced colitis mice were administered RIPK3 inhibitor (3 mg/ml) 3 times and their tissues were analyzed by immunohistochemistry. RIPK3, mixed lineage kinase domain-like (MLKL), phosphorylated MLKL, IL-17, and CD4 in colitis patient colon tissues were detected using confocal microscopy. Protein levels were measured using immunohistochemistry and ELISA. The differentiation of Th17 cells was evaluated using flow cytometry. The expression of proinflammatory cytokines and necroptosis in peripheral blood mononuclear cells from UC patients was decreased markedly by RIPK3 inhibitor treatment. We also observed that the injection of RIPK3 inhibitor improves colitis severity and protects intestinal destruction. RIPK3 inhibitor reduced necroptosis factors and proinflammatory cytokines in the colon and consequently protected colon devastation. The expression of inflammatory mediators in experimental colitis mice splenocytes was decreased significantly by RIPK3 inhibitor treatment. These results suggest that RIPK3 inhibitor ameliorates severity of experimental colitis and reduces inflammation through the inhibition of inflammatory response and necroptosis and support RIPK3-targeting substances for treatment of UC.
Collapse
Affiliation(s)
- Seung Hoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ji Ye Kwon
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jeonghyeon Moon
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - JeongWon Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jooyeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | | | - Keun-Hyung Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Om Darlami
- College of Pharmacy, Medical Campus, Gachon University, Incheon 21936, Korea
| | - Han Hee Lee
- Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Dong Yun Shin
- College of Pharmacy, Medical Campus, Gachon University, Incheon 21936, Korea
| | - Bo-In Lee
- Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.,Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
287
|
Bebber CM, Müller F, Prieto Clemente L, Weber J, von Karstedt S. Ferroptosis in Cancer Cell Biology. Cancers (Basel) 2020; 12:E164. [PMID: 31936571 PMCID: PMC7016816 DOI: 10.3390/cancers12010164] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
A major hallmark of cancer is successful evasion of regulated forms of cell death. Ferroptosis is a recently discovered type of regulated necrosis which, unlike apoptosis or necroptosis, is independent of caspase activity and receptor-interacting protein 1 (RIPK1) kinase activity. Instead, ferroptotic cells die following iron-dependent lipid peroxidation, a process which is antagonised by glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1). Importantly, tumour cells escaping other forms of cell death have been suggested to maintain or acquire sensitivity to ferroptosis. Therefore, therapeutic exploitation of ferroptosis in cancer has received increasing attention. Here, we systematically review current literature on ferroptosis signalling, cross-signalling to cellular metabolism in cancer and a potential role for ferroptosis in tumour suppression and tumour immunology. By summarising current findings on cell biology relevant to ferroptosis in cancer, we aim to point out new conceptual avenues for utilising ferroptosis in systemic treatment approaches for cancer.
Collapse
Affiliation(s)
- Christina M. Bebber
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
- Department I of Internal Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Fabienne Müller
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Laura Prieto Clemente
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Josephine Weber
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| | - Silvia von Karstedt
- Department of Translational Genomics, Medical Faculty, University of Cologne, Weyertal 155b, 50931 Cologne, Germany; (C.M.B.); (F.M.); (L.P.C.); (J.W.)
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), Medical Faculty, University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany
| |
Collapse
|
288
|
Ramraj SK, Elayapillai SP, Pelikan RC, Zhao YD, Isingizwe ZR, Kennedy AL, Lightfoot SA, Benbrook DM. Novel ovarian cancer maintenance therapy targeted at mortalin and mutant p53. Int J Cancer 2020; 147:1086-1097. [PMID: 31845320 DOI: 10.1002/ijc.32830] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/26/2019] [Accepted: 12/06/2019] [Indexed: 12/22/2022]
Abstract
Current ovarian cancer maintenance therapy is limited by toxicity and no proven impact on overall survival. To study a maintenance strategy targeted at missense mutant p53, we hypothesized that the release of mutant p53 from mortalin inhibition by the SHetA2 drug combined with reactivation of mutant p53 with the PRIMA-1MET drug inhibits growth and tumor establishment synergistically in a mutant-p53 dependent manner. The Cancer Genome Atlas (TCGA) data and serous ovarian tumors were evaluated for TP53 and HSPA9/mortalin status. SHetA2 and PRIMA-1MET were tested in ovarian cancer cell lines and fallopian tube secretory epithelial cells using isobolograms, fluorescent cytometry, Western blots and ELISAs. Drugs were administered to mice after peritoneal injection of MESOV mutant p53 ovarian cancer cells and prior to tumor establishment, which was evaluated by logistic regression. Fifty-eight percent of TP53 mutations were missense and there were no mortalin mutations in TCGA high-grade serous ovarian cancers. Mortalin levels were sequentially increased in serous benign, borderline and carcinoma tumors. SHetA2 caused p53 nuclear and mitochondrial accumulation in cancer, but not in healthy, cells. Endogenous or exogenous mutant p53 increased SHetA2 resistance. PRIMA-1MET decreased this resistance and interacted synergistically with SHetA2 in mutant and wild type p53-expressing cell lines in association with elevated reactive oxygen species/ATP ratios. Tumor-free rates in animals were 0% (controls), 25% (PRIMA1MET ), 42% (SHetA2) and 67% (combination). SHetA2 (p = 0.004) and PRIMA1MET (p = 0.048) functioned additively in preventing tumor development with no observed toxicity. These results justify the development of SHetA2 and PRIMA-1MET alone and in combination for ovarian cancer maintenance therapy.
Collapse
Affiliation(s)
- Satish K Ramraj
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Sugantha P Elayapillai
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Richard C Pelikan
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Yan D Zhao
- Biostatistics & Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Zitha R Isingizwe
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Amy L Kennedy
- Department of Pathology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Stanley A Lightfoot
- Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Doris M Benbrook
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Center for Cancer Prevention and Drug Development, Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK.,Obstetrics and Gynecologic, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
289
|
Identification of Two Kinase Inhibitors with Synergistic Toxicity with Low-Dose Hydrogen Peroxide in Colorectal Cancer Cells in vitro. Cancers (Basel) 2020; 12:cancers12010122. [PMID: 31906582 PMCID: PMC7016670 DOI: 10.3390/cancers12010122] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal carcinoma is among the most common types of cancers. With this disease, diffuse scattering in the abdominal area (peritoneal carcinosis) often occurs before diagnosis, making surgical removal of the entire malignant tissue impossible due to a large number of tumor nodules. Previous treatment options include radiation and its combination with intraperitoneal heat-induced chemotherapy (HIPEC). Both options have strong side effects and are often poor in therapeutic efficacy. Tumor cells often grow and proliferate dysregulated, with enzymes of the protein kinase family often playing a crucial role. The present study investigated whether a combination of protein kinase inhibitors and low-dose induction of oxidative stress (using hydrogen peroxide, H2O2) has an additive cytotoxic effect on murine, colorectal tumor cells (CT26). Protein kinase inhibitors from a library of 80 substances were used to investigate colorectal cancer cells for their activity, morphology, and immunogenicity (immunogenic cancer cell death, ICD) upon mono or combination. Toxic compounds identified in 2D cultures were confirmed in 3D cultures, and additive cytotoxicity was identified for the substances lavendustin A, GF109203X, and rapamycin. Toxicity was concomitant with cell cycle arrest, but except HMGB1, no increased expression of immunogenic markers was identified with the combination treatment. The results were validated for GF109203X and rapamycin but not lavendustin A in the 3D model of different colorectal (HT29, SW480) and pancreatic cancer cell lines (MiaPaca, Panc01). In conclusion, our in vitro data suggest that combining oxidative stress with chemotherapy would be conceivable to enhance antitumor efficacy in HIPEC.
Collapse
|
290
|
Yamazaki T, Vanpouille-Box C, Demaria S, Galluzzi L. Immunogenic Cell Death Driven by Radiation-Impact on the Tumor Microenvironment. Cancer Treat Res 2020; 180:281-296. [PMID: 32215874 DOI: 10.1007/978-3-030-38862-1_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunogenic cell death (ICD) is a particular form of cell death that can initiate adaptive immunity against antigens expressed by dying cells in the absence of exogenous adjuvants. This implies that cells undergoing ICD not only express antigens that are not covered by thymic tolerance, but also deliver adjuvant-like signals that enable the recruitment and maturation of antigen-presenting cells toward an immunostimulatory phenotype, culminating with robust cross-priming of antigen-specific CD8+ T cells. Such damage-associated molecular patterns (DAMPs), which encompass cellular proteins, small metabolites and cytokines, are emitted in a spatiotemporally defined manner in the context of failing adaptation to stress. Radiation therapy (RT) is a bona fide inducer of ICD, at least when employed according to specific doses and fractionation schedules. Here, we discuss the mechanisms whereby DAMPs emitted by cancer cells undergoing RT-driven ICD alter the functional configuration of the tumor microenvironment.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA.
- Université de Paris, Paris, France.
| |
Collapse
|
291
|
Farah BL, Yen PM, Koeberl DD. Links between autophagy and disorders of glycogen metabolism - Perspectives on pathogenesis and possible treatments. Mol Genet Metab 2020; 129:3-12. [PMID: 31787497 PMCID: PMC7836271 DOI: 10.1016/j.ymgme.2019.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 01/17/2023]
Abstract
The glycogen storage diseases are a group of inherited metabolic disorders that are characterized by specific enzymatic defects involving the synthesis or degradation of glycogen. Each disorder presents with a set of symptoms that are due to the underlying enzyme deficiency and the particular tissues that are affected. Autophagy is a process by which cells degrade and recycle unneeded or damaged intracellular components such as lipids, glycogen, and damaged mitochondria. Recent studies showed that several of the glycogen storage disorders have abnormal autophagy which can disturb normal cellular metabolism and/or mitochondrial function. Here, we provide a clinical overview of the glycogen storage disorders, a brief description of autophagy, and the known links between specific glycogen storage disorders and autophagy.
Collapse
Affiliation(s)
- Benjamin L Farah
- Department of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA..
| |
Collapse
|
292
|
Ashrafizadeh M, Tavakol S, Ahmadi Z, Roomiani S, Mohammadinejad R, Samarghandian S. Therapeutic effects of kaempferol affecting autophagy and endoplasmic reticulum stress. Phytother Res 2019; 34:911-923. [PMID: 31829475 DOI: 10.1002/ptr.6577] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 01/06/2023]
Abstract
Regulated cell death (RCD) guarantees to preserve organismal homeostasis. Apoptosis and autophagy are two major arms of RCD, while endoplasmic reticulum (ER) as a crucial organelle involved in proteostasis, promotes cells toward autophagy and apoptosis. Alteration in ER stress and autophagy machinery is responsible for a great number of diseases. Therefore, targeting those pathways appears to be beneficial in the treatment of relevant diseases. Meantime, among the traditional herb medicine, kaempferol as a flavonoid seems to be promising to modulate ER stress and autophagy and exhibits protective effects on malfunctioning cells. There are some reports indicating the capability of kaempferol in affecting autophagy and ER stress. In brief, kaempferol modulates autophagy in noncancerous cells to protect cells against malfunction, while it induces cell mortality derived from autophagy through the elevation of p-AMP-activated protein kinase, light chain-3-II, autophagy-related geness, and Beclin-1 in cancer cells. Noteworthy, kaempferol enhances cell survival through C/EBP homologous protein (CHOP) suppression and GRP78 increment in noncancerous cells, while it enhances cell mortality through the induction of unfolding protein response and CHOP increment in cancer cells. In this review, we discuss how kaempferol modulates autophagy and ER stress in noncancer and cancer cells to expand our knowledge of new pharmacological compounds for the treatment of associated diseases.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Sahar Roomiani
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
293
|
Abstract
Leishmaniases still represent a global scourge and new therapeutic tools are necessary to replace the current expensive, difficult to administer treatments that induce numerous adverse effects and for which resistance is increasingly worrying. In this context, the particularly original organization of the Leishmania parasite in comparison to higher eukaryotes is a great advantage. It allows for the development of new, very specific, and thus non-cytotoxic treatments. Among these originalities, Leishmania cell death can be cited. Despite a classic pattern of apoptosis, key mammalian apoptotic proteins are not present in Leishmania, such as caspases, cell death receptors, and anti-apoptotic molecules. Recent studies have helped to develop a better understanding of parasite cell death, identifying new proteins or even new apoptotic pathways. This review provides an overview of the current knowledge on Leishmania cell death, describing its physiological roles and its phenotype, and discusses the involvement of various proteins: endonuclease G, metacaspase, aquaporin Li-BH3AQP, calpains, cysteine proteinase C, LmjHYD36 and Lmj.22.0600. From these data, potential apoptotic pathways are suggested. This review also offers tools to identify new Leishmania cell death effectors. Lastly, different approaches to use this knowledge for the development of new therapeutic tools are suggested: either inhibition of Leishmania cell death or activation of cell death for instance by treating cells with proteins or peptides involved in parasite death fused to a cell permeant peptide or encapsulated into a lipidic vector to target intra-macrophagic Leishmania cells.
Collapse
Affiliation(s)
- Louise Basmaciyan
- UMR PAM A, Valmis Team, 2 rue Angélique Ducoudray, BP 37013, 21070 Dijon Cedex, France
| | - Magali Casanova
- Aix-Marseille University, CNRS, LISM, Institut de Microbiologie de la Méditerranée, 13402 Marseille Cedex 09, France
| |
Collapse
|
294
|
Ramos-Pérez C, Dominska M, Anaissi-Afonso L, Cazorla-Rivero S, Quevedo O, Lorenzo-Castrillejo I, Petes TD, Machín F. Cytological and genetic consequences for the progeny of a mitotic catastrophe provoked by Topoisomerase II deficiency. Aging (Albany NY) 2019; 11:11686-11721. [PMID: 31812950 PMCID: PMC6932922 DOI: 10.18632/aging.102573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/24/2019] [Indexed: 02/07/2023]
Abstract
Topoisomerase II (Top2) removes topological linkages between replicated chromosomes. Top2 inhibition leads to mitotic catastrophe (MC) when cells unsuccessfully try to split their genetic material between the two daughter cells. Herein, we have characterized the fate of these daughter cells in the budding yeast. Clonogenic and microcolony experiments, in combination with vital and apoptotic stains, showed that 75% of daughter cells become senescent in the short term; they are unable to divide but remain alive. Decline in cell vitality then occurred, yet slowly, uncoordinatedly when comparing pairs of daughters, and independently of the cell death mediator Mca1/Yca1. Furthermore, we showed that senescence can be modulated by ploidy, suggesting that gross chromosome imbalances during segregation may account for this phenotype. Indeed, we found that diploid long-term survivors of the MC are prone to genomic imbalances such as trisomies, uniparental disomies and terminal loss of heterozygosity (LOH), the latter affecting the longest chromosome arms.
Collapse
Affiliation(s)
- Cristina Ramos-Pérez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Escuela de Doctorado y Estudios de Postgrado, Universidad de La Laguna, Tenerife, Spain.,Present address: BenchSci Analytics Inc., Toronto, Canada
| | - Margaret Dominska
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Laura Anaissi-Afonso
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Escuela de Doctorado y Estudios de Postgrado, Universidad de La Laguna, Tenerife, Spain
| | - Sara Cazorla-Rivero
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Escuela de Doctorado y Estudios de Postgrado, Universidad de La Laguna, Tenerife, Spain
| | - Oliver Quevedo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Present address: Genomic Integrity Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Isabel Lorenzo-Castrillejo
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Thomas D Petes
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Félix Machín
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain.,Facultad de Ciencias de la Salud, Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
295
|
El-Maadawy WH, Hammam OA, Seif el-Din SH, El-Lakkany NM. α-Lipoic acid modulates liver fibrosis: A cross talk between TGF-β1, autophagy, and apoptosis. Hum Exp Toxicol 2019; 39:440-450. [DOI: 10.1177/0960327119891212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis are important players in the progression of hepatic fibrosis via activation of hepatic stellate cells (HSCs). Despite the recently depicted antifibrotic effects of alpha-lipoic acid (ALA), however, its modulatory effects on HSCs autophagy remain unverified. Our study aimed to elucidate the underlying antifibrotic mechanisms through which ALA mediates HSC autophagy and apoptosis. Liver fibrosis was induced via thioacetamide (TAA) intoxication in rats; TAA-intoxicated rats were treated with either silymarin or ALA. Effect of ALA on biochemical parameters and immunohistopathological examinations was measured and compared to silymarin. ALA restored normal hepatic architecture (S1 vs. S4), liver functions, hepatic glutathione, and transforming growth factor-β1 levels. ALA ameliorated hepatic levels of malondialdehyde, platelet-derived growth factor, tissue inhibitor metalloproteinases-1, hydroxyproline, and expression of alpha-smooth muscle actin. Moreover, ALA significantly reduced messenger RNA expression of LC3-II genes and triggered caspase-3 expression. Interestingly, ALA exhibited superior activities over silymarin regarding suppression of proliferation, activation and autophagy of HSCs, collagen deposition, and induction of HSCs apoptosis. In conclusion, treatment of TAA-intoxicated rats with ALA inhibited autophagy and induced apoptotic clearance of activated HSCs. Accordingly, this study provides mechanistic insights into the possible applicability of ALA in the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- WH El-Maadawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Giza, Egypt
| | - OA Hammam
- Department of Pathology, Theodor Bilharz Research Institute, Giza, Egypt
| | - SH Seif el-Din
- Department of Pharmacology, Theodor Bilharz Research Institute, Giza, Egypt
| | - NM El-Lakkany
- Department of Pharmacology, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
296
|
Raffinose increases autophagy and reduces cell death in UVB-irradiated keratinocytes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 201:111653. [DOI: 10.1016/j.jphotobiol.2019.111653] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/02/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023]
|
297
|
He D, Zheng J, Hu J, Chen J, Wei X. Long non-coding RNAs and pyroptosis. Clin Chim Acta 2019; 504:201-208. [PMID: 31794769 DOI: 10.1016/j.cca.2019.11.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/23/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
Long noncoding RNAs (lncRNAs) are defined as transcripts longer than 200 nucleotides that have no or only a low coding potential. They are involved in the progression of multiple diseases by the regulation of mechanisms related to epigenetic modifications and transcriptional and posttranscriptional processing. Recent studies have revealed an important function of lncRNAs in the regulation of pyroptosis, a type of programmed cell death associated with inflammatory responses that plays a critical role in many diseases. Through direct or indirect action on proteins related to the pyroptosis signaling pathway, lncRNAs are involved in the pathological processes related to cardiovascular diseases, kidney diseases, immune diseases and other diseases. Based on the expression characteristics of lncRNAs, this paper reviews the role of lncRNAs in regulating pyroptosis, aiming to provide new ideas for the research of lncRNAs regulating pyroptosis and treating pyroptosis-related diseases.
Collapse
Affiliation(s)
- Dong He
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China; School of Nursing, Xiangnan University, Chenzhou, Hunan 423000, China
| | - Jun Zheng
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421002, China
| | - Jia Hu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China
| | - Juan Chen
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China
| | - Xing Wei
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
298
|
Insights into mechanisms of pranoprofen-induced apoptosis and necroptosis in human corneal stromal cells. Toxicol Lett 2019; 320:9-18. [PMID: 31765691 DOI: 10.1016/j.toxlet.2019.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/13/2022]
Abstract
Pranoprofen (PPF) is a wildly used anti-inflammatory ophthalmic drug. It was reported that PPF could decrease early epithelialization of scrape wounds in rabbit cornea and could reduce cell activities of cultured human corneal endothelial cells. However, effects of PPF on corneal stromal cells playing important roles in corneal wound healing remain unknown. In this study,in vitro model of cultured human corneal stomal (HCS) cells and in vivo model of rabbit corneas were used to investigate the effects and underlying mechanisms of PPF. Our findings showed that high concentrations of PPF treatment (0.1 % to 0.0125 %) caused limited chromatin condensation and quickly decreased cell viability that was proved to initiate necroptosis in HCS cells through activating receptor interacting protein kinase (RIPK) and mixed lineage kinase domain-like (MLKL). While low concentrations of PPF treatment (0.00625 %) induced DNA fragmentation, apoptotic body formation, ROS generation, activation of caspases and increase in cytoplasmic content of Bad, Bax and cytoplasmic cytochrome c that suggested apoptosis happened through ROS-mediated caspase-dependent and caspase-independent pathways. Studies of rabbit corneas treated with 0.1 % PPF (the clinical concentration) showed that PPF could induce apoptosis of rabbit corneal stromal cells. This work would be helpful for better understanding cytotoxic effects PPF on human corneal cells.
Collapse
|
299
|
Colhado Rodrigues BL, Lallo MA, Perez EC. The Controversial Role of Autophagy in Tumor Development: A Systematic Review. Immunol Invest 2019; 49:386-396. [PMID: 31726897 DOI: 10.1080/08820139.2019.1682600] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is a natural regulatory mechanism of the cell that eliminates unnecessary and dysfunctional cellular components to maintain homeostasis. Several authors have demonstrated that this mechanism can be induced by pathological conditions as cancer. However, their role in tumor development is still a controversial issue in cancer research. Here, we discussed the most relevant findings concerning autophagy in tumor development. In this critical review performed with studies published between 2002 and 2018, we found that the main pathway involved in the autophagy process is the PI3K/AKT/mTOR intracellular signaling pathway. Regarding their role in cancer development, breast cancer is the main study target, followed by lung, prostate and colon cancer. In these issues, 46% of the works consulted suggesting that autophagy inhibits tumor progression by favor a better antitumor response, 4% suggest that favors growth and tumor progression and, 50% of the authors failed to establish whether autophagy inhibits or favors tumor development. Herein, we concluded that depending on the study model, autophagy may favor or inhibits growth and cancer progression.
Collapse
Affiliation(s)
- Bridilla Luiza Colhado Rodrigues
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista, São Paulo, Brazil.,Laboratório de Imunogenética, Instituto Butantan, São Paulo, Brazil
| | - Maria Anete Lallo
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista, São Paulo, Brazil
| | - Elizabeth Cristina Perez
- Programa de Pós-Graduação em Patologia Ambiental e Experimental, Universidade Paulista, São Paulo, Brazil
| |
Collapse
|
300
|
Bonora M, Wieckowski MR, Sinclair DA, Kroemer G, Pinton P, Galluzzi L. Targeting mitochondria for cardiovascular disorders: therapeutic potential and obstacles. Nat Rev Cardiol 2019; 16:33-55. [PMID: 30177752 DOI: 10.1038/s41569-018-0074-0] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A large body of evidence indicates that mitochondrial dysfunction has a major role in the pathogenesis of multiple cardiovascular disorders. Over the past 2 decades, extraordinary efforts have been focused on the development of agents that specifically target mitochondria for the treatment of cardiovascular disease. Despite such an intensive wave of investigation, no drugs specifically conceived to modulate mitochondrial functions are currently available for the clinical management of cardiovascular disease. In this Review, we discuss the therapeutic potential of targeting mitochondria in patients with cardiovascular disease, examine the obstacles that have restrained the development of mitochondria-targeting agents thus far, and identify strategies that might empower the full clinical potential of this approach.
Collapse
Affiliation(s)
- Massimo Bonora
- Ruth L. and David S. Gottesman Institute for Stem Cell, Regenerative Medicine Research, Department of Cell Biology and Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mariusz R Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA.,Department of Pharmacology, School of Medical Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Guido Kroemer
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Paolo Pinton
- Department of Morphology, Surgery, and Experimental Medicine, Section of Pathology, Oncology, and Experimental Biology, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy. .,Maria Cecilia Hospital, GVM Care & Research, E.S. Health Science Foundation, Cotignola, Italy.
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France. .,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA.
| |
Collapse
|