51
|
Targeting autophagy to overcome drug resistance: further developments. J Hematol Oncol 2020; 13:159. [PMID: 33239065 PMCID: PMC7687716 DOI: 10.1186/s13045-020-01000-2] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/15/2020] [Indexed: 12/13/2022] Open
Abstract
Inhibiting cell survival and inducing cell death are the main approaches of tumor therapy. Autophagy plays an important role on intracellular metabolic homeostasis by eliminating dysfunctional or unnecessary proteins and damaged or aged cellular organelles to recycle their constituent metabolites that enable the maintenance of cell survival and genetic stability and even promotes the drug resistance, which severely limits the efficacy of chemotherapeutic drugs. Currently, targeting autophagy has a seemingly contradictory effect to suppress and promote tumor survival, which makes the effect of targeting autophagy on drug resistance more confusing and fuzzier. In the review, we summarize the regulation of autophagy by emerging ways, the action of targeting autophagy on drug resistance and some of the new therapeutic approaches to treat tumor drug resistance by interfering with autophagy-related pathways. The full-scale understanding of the tumor-associated signaling pathways and physiological functions of autophagy will hopefully open new possibilities for the treatment of tumor drug resistance and the improvement in clinical outcomes.
Collapse
|
52
|
Sorrentino D, Frentzel J, Mitou G, Blasco RB, Torossian A, Hoareau-Aveilla C, Pighi C, Farcé M, Meggetto F, Manenti S, Espinos E, Chiarle R, Giuriato S. High Levels of miR-7-5p Potentiate Crizotinib-Induced Cytokilling and Autophagic Flux by Targeting RAF1 in NPM-ALK Positive Lymphoma Cells. Cancers (Basel) 2020; 12:cancers12102951. [PMID: 33066037 PMCID: PMC7650725 DOI: 10.3390/cancers12102951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Anaplastic lymphoma kinase positive anaplastic large cell lymphomas are a pediatric disease, which still needs treatment improvement. Crizotinib was the first ALK-targeted inhibitor used in clinics, but relapses are now known to occur. Current research efforts indicate that combined therapies could represent a superior strategy to eradicate malignant cells and prevent tumor recurrence. Autophagy is a self-digestion cellular process, known to be induced upon diverse cancer therapies. Our present work demonstrates that the potentiation of the crizotinib-induced autophagy flux, through the serine/threonine kinase RAF1 downregulation, drives ALK+ ALCL cells to death. These results should encourage further investigations on the therapeutic modulation of autophagy in this particular cancer settings and other ALK-related malignancies. Abstract Anaplastic lymphoma kinase positive anaplastic large cell lymphomas (ALK+ ALCL) are an aggressive pediatric disease. The therapeutic options comprise chemotherapy, which is efficient in approximately 70% of patients, and targeted therapies, such as crizotinib (an ALK tyrosine kinase inhibitor (TKI)), used in refractory/relapsed cases. Research efforts have also converged toward the development of combined therapies to improve treatment. In this context, we studied whether autophagy could be modulated to improve crizotinib therapy. Autophagy is a vesicular recycling pathway, known to be associated with either cell survival or cell death depending on the cancer and therapy. We previously demonstrated that crizotinib induced cytoprotective autophagy in ALK+ lymphoma cells and that its further intensification was associated with cell death. In line with these results, we show here that combined ALK and Rapidly Accelerated Fibrosarcoma 1 (RAF1) inhibition, using pharmacological (vemurafenib) or molecular (small interfering RNA targeting RAF1 (siRAF1) or microRNA-7-5p (miR-7-5p) mimics) strategies, also triggered autophagy and potentiated the toxicity of TKI. Mechanistically, we found that this combined therapy resulted in the decrease of the inhibitory phosphorylation on Unc-51-like kinase-1 (ULK1) (a key protein in autophagy initiation), which may account for the enforced autophagy and cytokilling effect. Altogether, our results support the development of ALK and RAF1 combined inhibition as a new therapeutic approach in ALK+ ALCL.
Collapse
Affiliation(s)
- Domenico Sorrentino
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Julie Frentzel
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Merck Serono S.A., Department of Biotechnology Process Sciences, Route de Fenil 25, Z.I. B, 1804 Corsier-sur-Vevey, Switzerland
| | - Géraldine Mitou
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Rafael B. Blasco
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
| | - Avédis Torossian
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Coralie Hoareau-Aveilla
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
| | - Chiara Pighi
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Manon Farcé
- Pôle Technologique du CRCT—Plateau de Cytométrie et Tri cellulaire—INSERM U1037, F-31037 Toulouse, France;
| | - Fabienne Meggetto
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Stéphane Manenti
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
| | - Estelle Espinos
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
| | - Roberto Chiarle
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Sylvie Giuriato
- Cancer Research Center of Toulouse, INSERM U1037—Université Toulouse III-Paul Sabatier—CNRS ERL5294, F-31037 Toulouse, France; (D.S.); (J.F.); (G.M.); (A.T.); (C.H.-A.); (F.M.); (S.M.); (E.E.)
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA; (R.B.B.); (C.P.); (R.C.)
- Ligue Nationale Contre le Cancer, équipe labellisée 2016, F-31037 Toulouse, France
- European Research Initiative on ALK-related malignancies (ERIA), Cambridge CB2 0QQ, UK
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain
- Correspondence: ; Tel.: +33-(5)-82-74-16-35
| |
Collapse
|
53
|
[Cdc37 Contributes to bortezomib resistance in multiple myeloma via autophagy]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:583-588. [PMID: 32810966 PMCID: PMC7449775 DOI: 10.3760/cma.j.issn.0253-2727.2020.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
目的 探索细胞分裂周期蛋白37(Cdc37)通过调控细胞自噬参与多发性骨髓瘤(MM)对硼替佐米(BTZ)耐药的机制,为MM的治疗提供新的思路。 方法 应用实时荧光定量PCR(qRT-PCR)和Western blot(WB)技术测定BTZ耐药细胞株ANBL-6.BR中Cdc37及自噬相关基因LC3b的表达水平。采用慢病毒转染技术上调ANBL-6.BR细胞中Cdc37的表达水平,WB技术检测LC3b的表达,流式细胞术检测BTZ诱导的细胞凋亡。采用shRNA干扰技术下调MM细胞株NCI-H929中Cdc37的表达水平,CCK-8法进一步验证Cdc37低表达与BTZ耐药相关,WB技术检测AKT/mTOR信号通路及自噬相关蛋白的表达变化;通过流式细胞术检测自噬抑制剂氯喹(CQ)是否可以增加耐药MM细胞对BTZ的敏感性。 结果 Cdc37在BTZ耐药细胞株ANBL-6.BR中低表达,而自噬相关基因LC3b在耐药株中高表达。上调ANBL-6.BR细胞中Cdc37的表达水平可抑制LC3b的表达,且增加了MM细胞对BTZ的敏感性。在MM细胞系NCI-H929中抑制Cdc37的表达导致BTZ耐药,同时引起自噬水平升高。而自噬抑制剂CQ可逆转Cdc37下调所引起的BTZ耐药。 结论 Cdc37可能通过调控细胞自噬参与MM细胞对BTZ的耐药过程。
Collapse
|
54
|
Autophagy as a decisive process for cell death. Exp Mol Med 2020; 52:921-930. [PMID: 32591647 PMCID: PMC7338414 DOI: 10.1038/s12276-020-0455-4] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/14/2020] [Accepted: 05/14/2020] [Indexed: 01/05/2023] Open
Abstract
Autophagy is an intracellular catabolic pathway in which cellular constituents are engulfed by autophagosomes and degraded upon autophagosome fusion with lysosomes. Autophagy serves as a major cytoprotective process by maintaining cellular homeostasis and recycling cytoplasmic contents. However, emerging evidence suggests that autophagy is a primary mechanism of cell death (autophagic cell death, ACD) and implicates ACD in several aspects of mammalian physiology, including tumor suppression and psychological disorders. However, little is known about the physiological roles and molecular mechanisms of ACD. In this review, we document examples of ACD and discuss recent progress in our understanding of its molecular mechanisms.
Collapse
|
55
|
Wang L, Tan Z, Zhang Y, Kady Keita N, Liu H, Zhang Y. ADAM12 silencing promotes cellular apoptosis by activating autophagy in choriocarcinoma cells. Int J Oncol 2020; 56:1162-1174. [PMID: 32319603 PMCID: PMC7115740 DOI: 10.3892/ijo.2020.5007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/17/2020] [Indexed: 12/14/2022] Open
Abstract
ADAM metallopeptidase domain 12 (ADAM12) has been demonstrated to mediate cell proliferation and apoptosis resistance in several types of cancer cells. However, the effect of ADAM12 silencing on the proliferation and apoptosis of choriocarcinoma cells remains unknown. The present study revealed that ADAM12 silencing significantly inhibited cellular activity and proliferation in the human choriocarcinoma JEG3 cell line and increased the rate of apoptosis. In addition, ADAM12 silencing significantly increased the expression levels of the autophagy proteins microtubule-associated protein-light-chain 3 (LC3B) and autophagy related 5 (ATG5) and the fluorescence density of LC3B in JEG-3 cells. However, the suppression of autophagy by 3-methyladenine could block ADAM12 silencing-induced cellular apoptosis. ADAM12 silencing reduced the levels of the inflammatory factors interleukin-1β, interferon-γ and TNF-α, and inactivated nuclear p65-NF-κB and p-mTOR in JEG-3 cells. The downregulation of p-mTOR expression by ADAM12 silencing was rescued in 3-methyladenine-treated JEG-3 cells, indicating that mTOR might participate in the autophagy-mediated pro-apoptotic effect of ADAM12 silencing. In conclusion, ADAM12 silencing promoted cellular apoptosis in human choriocarcinoma JEG3 cells, which might be associated with autophagy and the mTOR response. These findings indicate that ADAM12 silencing might be a potential novel therapeutic target for choriocarcinoma.
Collapse
Affiliation(s)
- Lin Wang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Zhihui Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ying Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Nankoria Kady Keita
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Huining Liu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
56
|
Wu H, Liu C, Yang Q, Xin C, Du J, Sun F, Zhou L. MIR145-3p promotes autophagy and enhances bortezomib sensitivity in multiple myeloma by targeting HDAC4. Autophagy 2020; 16:683-697. [PMID: 31242129 PMCID: PMC7138223 DOI: 10.1080/15548627.2019.1635380] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 01/04/2023] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with poor survival. Autophagy, a stress-responsive catabolic process mediated by lysosomal activity, plays a crucial role in the pathophysiology of MM. Growing evidence has indicated that dysregulated microRNAs (miRNAs) are associated with the aberrant autophagy in various human cancers. However, to date, few miRNAs have been reported to directly modulate autophagy in the pathobiology of MM. In this study, we investigated the role of MIR145-3p (microRNA 145-3p) in MM, with focus on cellular processes autophagy and cell death. Our results provided evidence that downregulation of MIR145-3p expression was associated with disease progression in human MM. MIR145-3p triggered autophagic flux through direct targeting of HDAC4 (histone deacetylase 4) in MM cells, leading to enhanced apoptosis. Silencing HDAC4 recapitulated the effects of MIR145-3p, whereas enforced expression of HDAC4 abrogated the effects of MIR145-3p. Furthermore, we showed that suppression of HDAC4 by MIR145-3p resulted in upregulation of the pro-apoptotic protein BCL2L11 and caused MTORC1 inactivation, which in turn led to enhanced autophagy and cell death. Importantly, we demonstrated that MIR145-3p mimic could potentiate the anti-MM activity of bortezomib in both in vitro and in vivo experiments. Overall, our findings indicate that MIR145-3p exerted a tumor suppression function in MM by inducing autophagic cell death and suggest that MIR145-3p-based targeted therapy would represent a novel strategy for MM treatment.Abbreviations: 3-MA: 3-methyladenine; 3'-UTR: 3'-untranslated region; 7-AAD: 7-aminoactinomycin D; ACTB: actin beta; ANXA5: annexin A5; ATG5: autophagy related 5; ATG7: autophagy related 7; B2M: beta-2-microglobulin; BAF: bafilomycin A1; BCL2L11: BCL2 like 11; Bort: bortezomib; CASP3: caspase 3; CCK-8: Cell Counting Kit-8; CQ: chloroquine; Ct: threshold cycle; ctrl: control; DAPI: 4',6-diamidino-2-phenylindole; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HDAC4: histone deacetylase 4; ISS: International Staging System; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; miRNAs: microRNAs; MIR145-3p: microRNA 145-3p; MM: multiple myeloma; mRNA: messenger RNA; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; PCs: plasma cells; PFS: progression-free survival; qRT-PCR: quantitative reverse transcription PCR; RPS6KB1: ribosomal protein S6 kinase B1; SD: standard deviation; siRNA: small interfering RNA; SQSTM1: sequestosome 1; STV: starvation; TUBB: tubulin beta class I.
Collapse
Affiliation(s)
- Hongkun Wu
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Chang Liu
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Qingyuan Yang
- Department of Clinical Laboratory Medicine, Tenth People’s Hospital of Tongji University, Shanghai, P.R. China
| | - Chengde Xin
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Juan Du
- Department of Hematology, The Myeloma & Lymphoma Center, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Fenyong Sun
- Department of Clinical Laboratory Medicine, Tenth People’s Hospital of Tongji University, Shanghai, P.R. China
| | - Lin Zhou
- Department of Laboratory Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, P.R. China
| |
Collapse
|
57
|
Ishaq M, Ojha R, Sharma AP, Singh SK. Autophagy in cancer: Recent advances and future directions. Semin Cancer Biol 2020; 66:171-181. [PMID: 32201367 DOI: 10.1016/j.semcancer.2020.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 02/10/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is being explored as a potential therapeutic target for enhancing the cytotoxic effects of chemotherapeutic regimens in various malignancies. Autophagy plays a very important role in cancer pathogenesis. Here, we discuss the updates on the modulation of autophagy via dynamic interactions with different organelles and the exploitation of selective autophagy for exploring therapeutic strategies. We further discuss the role of autophagy inhibitors in cancer preclinical and clinical trials, novel autophagy inhibitors, and challenges likely to be faced by clinicians while inducting autophagy modulators in clinical practice.
Collapse
Affiliation(s)
- Mohd Ishaq
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Rani Ojha
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Aditya P Sharma
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Shrawan K Singh
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
58
|
Smyth P, Sessler T, Scott CJ, Longley DB. FLIP(L): the pseudo-caspase. FEBS J 2020; 287:4246-4260. [PMID: 32096279 PMCID: PMC7586951 DOI: 10.1111/febs.15260] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/10/2020] [Accepted: 02/24/2020] [Indexed: 12/27/2022]
Abstract
Possessing structural homology with their active enzyme counterparts but lacking catalytic activity, pseudoenzymes have been identified for all major enzyme groups. Caspases are a family of cysteine‐dependent aspartate‐directed proteases that play essential roles in regulating cell death and inflammation. Here, we discuss the only human pseudo‐caspase, FLIP(L), a paralog of the apoptosis‐initiating caspases, caspase‐8 and caspase‐10. FLIP(L) has been shown to play a key role in regulating the processing and activity of caspase‐8, thereby modulating apoptotic signaling mediated by death receptors (such as TRAIL‐R1/R2), TNF receptor‐1 (TNFR1), and Toll‐like receptors. In this review, these canonical roles of FLIP(L) are discussed. Additionally, a range of nonclassical pseudoenzyme roles are described, in which FLIP(L) functions independently of caspase‐8. These nonclassical pseudoenzyme functions enable FLIP(L) to play key roles in the regulation of a wide range of biological processes beyond its canonical roles as a modulator of cell death.
Collapse
Affiliation(s)
- Peter Smyth
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Tamas Sessler
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Christopher J Scott
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| | - Daniel B Longley
- The Patrick G Johnston Centre for Cancer Research, Queen's University, Belfast, UK
| |
Collapse
|
59
|
Bclaf1 is a direct target of HIF-1 and critically regulates the stability of HIF-1α under hypoxia. Oncogene 2020; 39:2807-2818. [PMID: 32029898 DOI: 10.1038/s41388-020-1185-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/10/2019] [Accepted: 01/23/2020] [Indexed: 01/04/2023]
Abstract
Hypoxic stress is intimately connected with tumor progression, with hypoxia-inducible factor-1α (HIF-1α) being a critical regulator in this process. HIF-1α is stabilized in response to hypoxia, which is required for the induction of gene transcriptions important for hypoxic adaptation. Bclaf1 is a multifunctional protein involved in tumorigenesis, however, its role in this process is not well characterized. Here we report Bclaf1 is a direct transcriptional target of HIF-1 and upregulated in multiple cell lines during hypoxia. Importantly, we found Bclaf1 is involved in the stabilization of HIF-1α during long-term hypoxic treatments. Compared with the control cells, the protein level and stability of HIF-1α in Bclaf1 knockdown or knockout cells is greatly compromised after long-term hypoxic treatments, concomitant with the impaired inductions of HIF-1 target gene transcription. Bclaf1 knockout HeLa cells exhibit a reduced tumor growth in mice xenografts, in which the expressions of HIF-1α and its target genes are also decreased. Bclaf1 binds to HIF-1α in the nucleus, and this interaction is required for Bclaf1 to stabilize HIF-1α in hypoxic condition. These results uncover a positive feedback loop, HIF-1-Bclaf1, that sustains HIF-1 activity during long-term hypoxic conditions by binding to and protecting HIF-1α from degradation, and suggest that Bclaf1 may promote tumor progression by enhancing HIF-1α stability.
Collapse
|
60
|
Abstract
Caspases are a family of conserved cysteine proteases that play key roles in programmed cell death and inflammation. In multicellular organisms, caspases are activated via macromolecular signaling complexes that bring inactive procaspases together and promote their proximity-induced autoactivation and proteolytic processing. Activation of caspases ultimately results in programmed execution of cell death, and the nature of this cell death is determined by the specific caspases involved. Pioneering new research has unraveled distinct roles and cross talk of caspases in the regulation of programmed cell death, inflammation, and innate immune responses. In-depth understanding of these mechanisms is essential to foster the development of precise therapeutic targets to treat autoinflammatory disorders, infectious diseases, and cancer. This review focuses on mechanisms governing caspase activation and programmed cell death with special emphasis on the recent progress in caspase cross talk and caspase-driven gasdermin D-induced pyroptosis.
Collapse
Affiliation(s)
- Sannula Kesavardhana
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; , ,
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA; , ,
| | | |
Collapse
|
61
|
Datan E, Salman S. Autophagic cell death in viral infection: Do TAM receptors play a role? TAM RECEPTORS IN HEALTH AND DISEASE 2020; 357:123-168. [DOI: 10.1016/bs.ircmb.2020.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
62
|
Zheng Z, Wang L, Cheng S, Wang Y, Zhao W. Autophagy and Myeloma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:625-631. [PMID: 32671780 DOI: 10.1007/978-981-15-4272-5_45] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Multiple myeloma is a hematological malignancy. It is characterized by the abnormal clonal proliferation of malignant plasma cells in the bone marrow and the secretion of a large number of monoclonal immunoglobulins or light chains, causing bone destruction, elevated blood calcium levels, anemia, and renal dysfunction. Autophagy has a dual role in the autophagy of myeloma cells. On the one hand, autophagy eliminates abnormal proteins and organelles in cells, prevents gene damage, and inhibits tumorigenesis. On the other hand, once tumors are formed, tumor cells use autophagy to ensure their survival under nutrient-deficient and hypoxic conditions. Excessive autophagy promotes another form of death in tumor cells, autophagic cell death. Targeted autophagy is becoming another new myeloma treatment strategy.
Collapse
Affiliation(s)
- Zhong Zheng
- State Key Laboratory of Medical Genomics, Shanghai Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- State Key Laboratory of Medical Genomics, Shanghai Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Cheng
- State Key Laboratory of Medical Genomics, Shanghai Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- State Key Laboratory of Medical Genomics, Shanghai Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Zhao
- State Key Laboratory of Medical Genomics, Shanghai Rui Jin Hospital, Shanghai Institute of Hematology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
63
|
Liao Y, Duan B, Zhang Y, Zhang X, Xia B. Excessive ER-phagy mediated by the autophagy receptor FAM134B results in ER stress, the unfolded protein response, and cell death in HeLa cells. J Biol Chem 2019; 294:20009-20023. [PMID: 31748416 PMCID: PMC6937584 DOI: 10.1074/jbc.ra119.008709] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Autophagy is typically a prosurvival cellular process that promotes the turnover of long-lived proteins and damaged organelles, but it can also induce cell death. We have previously reported that the small molecule Z36 induces autophagy along with autophagic cell death in HeLa cells. In this study, we analyzed differential gene expression in Z36-treated HeLa cells and found that Z36-induced endoplasmic reticulum-specific autophagy (ER-phagy) results in ER stress and the unfolded protein response (UPR). This result is in contrast to the common notion that autophagy is generally activated in response to ER stress and the UPR. We demonstrate that Z36 up-regulates the expression levels of FAM134B, LC3, and Atg9, which together mediate excessive ER-phagy, characterized by forming increased numbers of autophagosomes with larger sizes. We noted that the excessive ER-phagy accelerates ER degradation and impairs ER homeostasis and thereby triggers ER stress and the UPR as well as ER-phagy-dependent cell death. Interestingly, overexpression of FAM134B alone in HeLa cells is sufficient to impair ER homeostasis and cause ER stress and cell death. These findings suggest a mechanism involving FAM134B activity for ER-phagy to promote cell death.
Collapse
Affiliation(s)
- Yangjie Liao
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Bo Duan
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yufei Zhang
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | | - Bin Xia
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
64
|
BCLAF1 promotes cell proliferation, invasion and drug-resistance though targeting lncRNA NEAT1 in hepatocellular carcinoma. Life Sci 2019; 242:117177. [PMID: 31870774 DOI: 10.1016/j.lfs.2019.117177] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/11/2023]
Abstract
AIMS In the present research, we aimed to investigate the effect of Bcl-2-associated transcription factor 1 (BCLAF1) on hepatocellular carcinoma and further explore the special molecular mechanism. MAIN METHODS The expression of BCLAF1 was analyzed in tumor tissues and different hepatocellular cancer cell lines by real-time RT-PCR and Western blot. Cell proliferation and invasion was explored using MTT and Transwell assay respectively. In addition, luciferase reporter assay was performed to determine the binding activity of BCLAF1 and Nuclear enrichment-rich transcription factor 1 (NEAT1) promoter. Finally, the IC50 for 5-Fluorouracil (5-Fu) was measured by MTT assay, and Western blot was used to determine the expression of P-glycoprotein (P-gp) and multidrug resistance protein1 (MRP1). KEY FINDING The result revealed that BCLAF1 was highly expressed in hepatocellular carcinoma tissues and cells. In addition, BCLAF1-siRNA inhibited the proliferation and invasion of hepatocellular carcinoma cells, and overexpression of BCLAF1 promoted proliferation and invasion. Furthermore, luciferase reporter assay demonstrated that BCLAF1 directly interact with lncNEAT1 promoter and improved NEAT1 expression, and BCLAF1 promoted proliferation and invasion through targeting lncRNA NEAT1. What's more, BCLAF1 promoted 5-Fu resistance and the expression of P-gp and MRP1 in hepatocellular carcinoma cells by targeting NEAT1. SIGNIFICANCE The results of the present study suggested that BCLAF1 might be a new gene related to proliferation and drug-resistance of hepatocellular carcinoma. In the future, the search for a deep and reasonable mechanism for the role of BCLAF1 will help us to understand its function more comprehensively, and finally find a new method for the treatment of human cancer.
Collapse
|
65
|
Van Opdenbosch N, Lamkanfi M. Caspases in Cell Death, Inflammation, and Disease. Immunity 2019; 50:1352-1364. [PMID: 31216460 DOI: 10.1016/j.immuni.2019.05.020] [Citation(s) in RCA: 820] [Impact Index Per Article: 136.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023]
Abstract
Caspases are an evolutionary conserved family of cysteine proteases that are centrally involved in cell death and inflammation responses. A wealth of foundational insight into the molecular mechanisms that control caspase activation has emerged in recent years. Important advancements include the identification of additional inflammasome platforms and pathways that regulate activation of inflammatory caspases; the discovery of gasdermin D as the effector of pyroptosis and interleukin (IL)-1 and IL-18 secretion; and the existence of substantial crosstalk between inflammatory and apoptotic initiator caspases. A better understanding of the mechanisms regulating caspase activation has supported initial efforts to modulate dysfunctional cell death and inflammation pathways in a suite of communicable, inflammatory, malignant, metabolic, and neurodegenerative diseases. Here, we review current understanding of caspase biology with a prime focus on the inflammatory caspases and outline important topics for future experimentation.
Collapse
Affiliation(s)
- Nina Van Opdenbosch
- Janssen Immunosciences, World Without Disease Accelerator, Pharmaceutical Companies of Johnson & Johnson, Beerse, 2340, Belgium
| | - Mohamed Lamkanfi
- Janssen Immunosciences, World Without Disease Accelerator, Pharmaceutical Companies of Johnson & Johnson, Beerse, 2340, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, 9000, Belgium.
| |
Collapse
|
66
|
Yu S, Wang X, Dou N, Zhou J, Gao Y, Li Y. B-cell lymphoma-2-associated transcription factor 1 is overexpressed and contributes to sorafenib resistance in hepatocellular carcinoma. Hepatol Res 2019; 49:1329-1340. [PMID: 31237058 DOI: 10.1111/hepr.13395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/16/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
AIM B-cell lymphoma-2-associated transcription factor 1 (BCLAF1) is involved in various biological processes including tumorigenesis, but its function and expression in hepatocellular carcinoma (HCC) is little known, and its clinical value in HCC has not yet been defined. METHODS The protein level of BCLAF1 in HCC specimens and paired adjacent normal tissues was examined by immunohistochemical staining. The effects of BCLAF1 on autophagy in HCC cells were detected by confocal microscopy, transmission electron microscopy, and western blot analysis. Cell proliferation and tumorigenicity assays were carried out in vitro and in vivo. Flow cytometry assay was used to determine the apoptosis level of HCC cells. The correlation of BCLAF1 and sorafenib resistance in HCC was analyzed by the Kaplan-Meier survival method. RESULTS High expression of BCLAF1 was found in HCC tissues compared with adjacent normal tissues, and higher BCLAF1 expression was correlated with higher tumor-node-metastasis stage, worse differentiation, and worse prognosis of HCC patients. BCLAF1 could induce autophagy in HCC cells in response to starvation and BCLAF1-mediated autophagy could enhance cell proliferation and impede cell apoptosis under stress conditions. Animal experiments indicated that BCLAF1 promoted tumorigenicity of HCC cells in vivo. More importantly, high expression of BCLAF1 might contribute to sorafenib resistance in HCC patients. CONCLUSIONS BCLAF1 is a potential oncogene in HCC by inducing autophagy to maintain tumor cell growth in response to stress conditions, and it could serve as a potential biomarker for predicting the prognosis of HCC patients and screening patients who are suitable for sorafenib therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yandong Li
- Department of Oncology.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
67
|
CD19-positive antibody-secreting cells provide immune memory. Blood Adv 2019; 2:3163-3176. [PMID: 30478153 DOI: 10.1182/bloodadvances.2017015172] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
Long-lived antibody-secreting cells (ASCs) are critical for the maintenance of humoral immunity through the continued production of antibodies specific for previously encountered pathogen or vaccine antigens. Recent reports describing humoral immune memory have suggested the importance of long-lived CD19- bone marrow (BM) ASCs, which secrete antibodies recognizing previously encountered vaccine antigens. However, these reports do not agree upon the unique contribution of the CD19+ BM ASC subset toward humoral immunity. Here, we found both CD19+ and negative ASCs from human BM were similar in functional capacity to react to a number of vaccine antigens via ELISpot assays. The CD19+ cells were the predominant ASC population found in lymphoid tissues, and unlike the CD19- ASCs, which were found only in spleen and BM, the CD19+ ASCs were found in tonsil and blood. CD19+ ASCs from the BM, spleen, and tonsil were capable of recognizing polio vaccine antigens, indicating the CD19+ ASC cells play a novel role in long-lasting immune defense. Comparative gene expression analysis indicated CD19+ and negative BM ASCs differed significantly by only 14 distinct messenger RNAs and exhibited similar gene expression for cell cycle, autophagy, and apoptosis control necessary for long life. In addition, we show identical CDR-H3 sequences found on both BM ASC subsets, indicating a shared developmental path. Together, these results provide novel insight for the distribution, function, genetic regulation, and development of long-lived ASCs and may not only impact improved cell therapies but also enhance strategies for vaccine development.
Collapse
|
68
|
Xu SJ, Zhang F, Wang LJ, Hao MH, Yang XJ, Li NN, Ji HL, Xu P. Flavonoids of Rosa roxburghii Tratt offers protection against radiation induced apoptosis and inflammation in mouse thymus. Apoptosis 2019; 23:470-483. [PMID: 29995207 DOI: 10.1007/s10495-018-1466-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The present study evaluated the protective effect of the natural compound flavonoids of Rosa roxburghii Tratt (FRT) against γ-radiation-induced apoptosis and inflammation in mouse thymus cells in vivo and in vitro. Thymus cells and mice were exposed to 60Co γ-ray at a dose of 6 Gy. The radiation treatment induced significant cell apoptosis and inflammation. Radiation increased the expressions of cleaved caspase 3/8-10, AIF, and PARP-1, and FRT could mitigate their activation and inhibit subsequent apoptosis in the thymus both in vitro or in vivo. Irradiation increased the mRNA expression of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-κB. Our results also indicated that FRT alleviated gene expression of some inflammatory factors such as ICAM-1/VCAM-1, TNF-α/NF-κB, but not IL-1α/IL-6. Irradiation increased the protein expression levels of ICAM-1/VCAM-1, IL-1α/IL-6 and TNF-α/NF-Κb, and our results also indicated that FRT alleviated protein level expression of certain inflammatory factors such as ICAM-1, IL-1α/IL-6, TNF-α/NF-κB, but not VCAM-1. Our results suggested that FRT enhanced radioprotection at least partially by regulating caspase 3/8-10, AIF, and PARP-1 to reduce apoptosis and by regulating ICAM-1, IL-1α/IL-6, TNF-α/NF-κB to reduce inflammation.
Collapse
Affiliation(s)
- Sai-Juan Xu
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Fan Zhang
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Li-Juan Wang
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Ming-Hua Hao
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Xian-Jun Yang
- International Joint Research Laboratory for Recombiant Pharmaceutical Protein Expression System of Henan, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Na-Na Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Hong-Long Ji
- Department of Cellular and Molecular Biology, University of Texas Health Northeast, Tyler, TX, 75708, USA. .,Institute of Lung and Molecular Therapy, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| | - Ping Xu
- Department of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, Henan, China.
| |
Collapse
|
69
|
Caspase-10 inhibits ATP-citrate lyase-mediated metabolic and epigenetic reprogramming to suppress tumorigenesis. Nat Commun 2019; 10:4255. [PMID: 31534141 PMCID: PMC6751159 DOI: 10.1038/s41467-019-12194-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/27/2019] [Indexed: 01/18/2023] Open
Abstract
Caspase-10 belongs to the class of initiator caspases and is a close homolog of caspase-8. However, the lack of caspase-10 in mice and limited substrate repertoire restricts the understanding of its physiological functions. Here, we report that ATP-citrate lyase (ACLY) is a caspase-10 substrate. Caspase-10 cleaves ACLY at the conserved Asp1026 site under conditions of altered metabolic homeostasis. Cleavage of ACLY abrogates its enzymatic activity and suppresses the generation of acetyl-CoA, which is critical for lipogenesis and histone acetylation. Thus, caspase-10-mediated ACLY cleavage results in reduced intracellular lipid levels and represses GCN5-mediated histone H3 and H4 acetylation. Furthermore, decline in GCN5 activity alters the epigenetic profile, resulting in downregulation of proliferative and metastatic genes. Thus caspase-10 suppresses ACLY-promoted malignant phenotype. These findings expand the substrate repertoire of caspase-10 and highlight its pivotal role in inhibiting tumorigenesis through metabolic and epigenetic mechanisms. Caspases are most closely associated with cell death, but many have other cellular functions. Here, Das et al. find that upon metabolic stress, caspase-10 cleaves ACLY to regulate metabolic homeostasis and epigenetic reprogramming by altering Acetyl-CoA levels.
Collapse
|
70
|
Wang Z, Ni F, Yu F, Cui Z, Zhu X, Chen J. Prognostic significance of mRNA expression of CASPs in gastric cancer. Oncol Lett 2019; 18:4535-4554. [PMID: 31611962 PMCID: PMC6781674 DOI: 10.3892/ol.2019.10816] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022] Open
Abstract
Current studies suggest that the cysteinyl aspartate specific proteinase (caspase/CASP) family may be closely associated with apoptosis. Scientists have suggested that caspases may be a key to the development of more effective anti-cancer therapies. However, the prognostic value of CASP expression in gastric cancer (GC) remains unclear. Using a Kaplan-Meier plotter online database, the predictive prognostic significance of the expression of 12 CASPs genes (CASP1, CASP2, CASP3, CASP4, CASP5, CASP6, CASP7, CASP8, CASP9, CASP10, CASP12 and CASP14) to overall survival (OS) in different clinicopathological features, including Lauren classification, pathological stages, therapies employed and differentiation in gastric cancer patients was explored. The present study revealed that higher CASP1, 2, 3, 4, 5, 6, 7 and 8 mRNA expression was associated with better OS, whereas higher expression of CASP9, 10, 12 and 14 showed an unfavorable OS in all GC patients. Moreover, CASP1 to 8 were all associated with favorable OS in intestinal type and diffuse type classified by Lauren classification. Therefore, the results of the present study suggested that the CASP family may function as new prognostic indicators in GC and may be helpful in making treatment decisions.
Collapse
Affiliation(s)
- Zixiang Wang
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fubiao Ni
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fangyi Yu
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhonghui Cui
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiandong Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jicai Chen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
71
|
Carrino M, Quotti Tubi L, Fregnani A, Canovas Nunes S, Barilà G, Trentin L, Zambello R, Semenzato G, Manni S, Piazza F. Prosurvival autophagy is regulated by protein kinase CK1 alpha in multiple myeloma. Cell Death Discov 2019; 5:98. [PMID: 31123604 PMCID: PMC6529432 DOI: 10.1038/s41420-019-0179-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/09/2019] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is a tumor of plasma cells (PCs). Due to the intense immunoglobulin secretion, PCs are prone to endoplasmic reticulum stress and activate several stress-managing pathways, including autophagy. Indeed, autophagy deregulation is maladaptive for MM cells, resulting in cell death. CK1α, a pro-survival kinase in MM, has recently been involved as a regulator of the autophagic flux and of the transcriptional competence of the autophagy-related transcription factor FOXO3a in several cancers. In this study, we investigated the role of CK1α in autophagy in MM. To study the autophagic flux we generated clones of MM cell lines expressing the mCherry-eGFP-LC3B fusion protein. We observed that CK1 inhibition with the chemical ATP-competitive CK1 α/δ inhibitor D4476 resulted in an impaired autophagic flux, likely due to an alteration of lysosomes acidification. However, D4476 caused the accumulation of the transcription factor FOXO3a in the nucleus, and this was paralleled by the upregulation of mRNA coding for autophagic genes. Surprisingly, silencing of CK1α by RNA interference triggered the autophagic flux. However, FOXO3a did not shuttle into the nucleus and the transcription of autophagy-related FOXO3a-dependent genes was not observed. Thus, while the chemical inhibition with the dual CK1α/δ inhibitor D4476 induced cell death as a consequence of an accumulation of ineffective autophagic vesicles, on the opposite, CK1α silencing, although it also determined apoptosis, triggered a full activation of the early autophagic flux, which was then not supported by the upregulation of autophagic genes. Taken together, our results indicate that the family of CK1 kinases may profoundly influence MM cells survival also through the modulation of the autophagic pathway.
Collapse
Affiliation(s)
- Marilena Carrino
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Laura Quotti Tubi
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Anna Fregnani
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sara Canovas Nunes
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy.,Boston Children's Hospital/Harvard Medical School, Boston, MA USA
| | - Gregorio Barilà
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Livio Trentin
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Renato Zambello
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- 1Department of Medicine, Hematology and Clinical Immunology Branch, University of Padova, Padova, Italy.,2Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
72
|
Janker L, Mayer RL, Bileck A, Kreutz D, Mader JC, Utpatel K, Heudobler D, Agis H, Gerner C, Slany A. Metabolic, Anti-apoptotic and Immune Evasion Strategies of Primary Human Myeloma Cells Indicate Adaptations to Hypoxia. Mol Cell Proteomics 2019; 18:936-953. [PMID: 30792264 PMCID: PMC6495257 DOI: 10.1074/mcp.ra119.001390] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Indexed: 12/26/2022] Open
Abstract
Multiple Myeloma (MM) is an incurable plasma cell malignancy primarily localized within the bone marrow (BM). It develops from a premalignant stage, monoclonal gammopathy of undetermined significance (MGUS), often via an intermediate stage, smoldering MM (SMM). The mechanisms of MM progression have not yet been fully understood, all the more because patients with MGUS and SMM already carry similar initial mutations as found in MM cells. Over the last years, increased importance has been attributed to the tumor microenvironment and its role in the pathophysiology of the disease. Adaptations of MM cells to hypoxic conditions in the BM have been shown to contribute significantly to MM progression, independently from the genetic predispositions of the tumor cells. Searching for consequences of hypoxia-induced adaptations in primary human MM cells, CD138-positive plasma cells freshly isolated from BM of patients with different disease stages, comprising MGUS, SMM, and MM, were analyzed by proteome profiling, which resulted in the identification of 6218 proteins. Results have been made fully accessible via ProteomeXchange with identifier PXD010600. Data previously obtained from normal primary B cells were included for comparative purposes. A principle component analysis revealed three clusters, differentiating B cells as well as MM cells corresponding to less and more advanced disease stages. Comparing these three clusters pointed to the alteration of pathways indicating adaptations to hypoxic stress in MM cells on disease progression. Protein regulations indicating immune evasion strategies of MM cells were determined, supported by immunohistochemical staining, as well as transcription factors involved in MM development and progression. Protein regulatory networks related to metabolic adaptations of the cells became apparent. Results were strengthened by targeted analyses of a selected panel of metabolites in MM cells and MM-associated fibroblasts. Based on our data, new opportunities may arise for developing therapeutic strategies targeting myeloma disease progression.
Collapse
Affiliation(s)
- Lukas Janker
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Rupert L Mayer
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Andrea Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dominique Kreutz
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Johanna C Mader
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Kirsten Utpatel
- Department of Pathology, University Regensburg, Regensburg, Germany
| | - Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Hermine Agis
- Department of Oncology, University Clinic for Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria;.
| |
Collapse
|
73
|
Safa AR, Kamocki K, Saadatzadeh MR, Bijangi-Vishehsaraei K. c-FLIP, a Novel Biomarker for Cancer Prognosis, Immunosuppression, Alzheimer's Disease, Chronic Obstructive Pulmonary Disease (COPD), and a Rationale Therapeutic Target. BIOMARKERS JOURNAL 2019; 5:4. [PMID: 32352084 PMCID: PMC7189798 DOI: 10.36648/2472-1646.5.1.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dysregulation of c-FLIP (cellular FADD-like IL-1β-converting enzyme inhibitory protein) has been shown in several diseases including cancer, Alzheimer's disease, and chronic obstructive pulmonary disease (COPD). c-FLIP is a critical anti-cell death protein often overexpressed in tumors and hematological malignancies and its increased expression is often associated with a poor prognosis. c-FLIP frequently exists as long (c-FLIPL) and short (c-FLIPS) isoforms, regulates its anti-cell death functions through binding to FADD (FAS associated death domain protein), an adaptor protein known to activate caspases-8 and -10 and links c-FLIP to several cell death regulating complexes including the death-inducing signaling complex (DISC) formed by various death receptors. c-FLIP also plays a critical role in necroptosis and autophagy. Furthermore, c-FLIP is able to activate several pathways involved in cytoprotection, proliferation, and survival of cancer cells through various critical signaling proteins. Additionally, c-FLIP can inhibit cell death induced by several chemotherapeutics, anti-cancer small molecule inhibitors, and ionizing radiation. Moreover, c-FLIP plays major roles in aiding the survival of immunosuppressive tumor-promoting immune cells and functions in inflammation, Alzheimer's disease (AD), and chronic obstructive pulmonary disease (COPD). Therefore, c-FLIP can serve as a versatile biomarker for cancer prognosis, a diagnostic marker for several diseases, and an effective therapeutic target. In this article, we review the functions of c-FLIP as an anti-apoptotic protein and negative prognostic factor in human cancers, and its roles in resistance to anticancer drugs, necroptosis and autophagy, immunosuppression, Alzheimer's disease, and COPD.
Collapse
Affiliation(s)
- Ahmad R Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - Krzysztof Kamocki
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, USA
| | - M Reza Saadatzadeh
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, USA
| | | |
Collapse
|
74
|
Zhou X, Wen Y, Tian Y, He M, Ke X, Huang Z, He Y, Liu L, Scharf A, Lu M, Zhang G, Deng Y, Yan Y, Mayer MP, Chen X, Zou F. Heat Shock Protein 90α-Dependent B-Cell-2-Associated Transcription Factor 1 Promotes Hepatocellular Carcinoma Proliferation by Regulating MYC Proto-Oncogene c-MYC mRNA Stability. Hepatology 2019; 69:1564-1581. [PMID: 30015413 PMCID: PMC6586158 DOI: 10.1002/hep.30172] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/20/2018] [Indexed: 12/14/2022]
Abstract
B-cell lymphoma 2 (Bcl-2)-associated transcription factor 1 (Bclaf1) is known to be involved in diverse biological processes, but, to date, there has been no evidence for any functional role of Bclaf1 in hepatocellular carcinoma (HCC) progression. Here, we demonstrate that Bclaf1 is frequently up-regulated in HCC and that Bclaf1 up-regulation is associated with Edmondson grade, lower overall survival rates, and poor prognosis. Overexpression of Bclaf1 in HCC cell lines HepG2 and Huh7 promoted proliferation considerably, whereas Bclaf1 knockdown had the opposite effect. Xenograft tumors grown from Bclaf1 knockdown Huh7 cells had smaller tumor volumes than tumors grown from control cells. Furthermore, our study describes MYC proto-oncogene (c-Myc) as a downstream target of Bclaf1, given that Bclaf1 regulates c-MYC expression posttranscriptionally by its RS domain. To exert this function, Bclaf1 must interact with the molecular chaperone, heat shock protein 90 alpha (Hsp90α). In HCC tissue samples, Hsp90α levels were also increased significantly and Hsp90α-Bclaf1 interaction was enhanced. Bclaf1 interacts with the C-terminal domain of Hsp90α, and this interaction is disrupted by the C-terminal domain inhibitor, novobiocin (NB), resulting in proteasome-dependent degradation of Bclaf1. Moreover, NB-induced disruption of Hsp90α-Bclaf1 interaction dampened the production of mature c-MYC mRNA and attenuated tumor cell growth in vitro and in vivo. Conclusion: Our findings suggest that Bclaf1 affects HCC progression by manipulating c-MYC mRNA stability and that the Hsp90α/Bclaf1/c-Myc axis might be a potential target for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Xueqiong Zhou
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Ying Wen
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Ye Tian
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Meiling He
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Xiangyu Ke
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Zhizhou Huang
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Yangfan He
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Lixia Liu
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Annette Scharf
- Center for Molecular Biology of Heidelberg University (ZMBH)DKFZ‐ZMBH‐AllianceHeidelbergGermany
| | - Meiting Lu
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Guowei Zhang
- Department of Hepatobiliary SurgeryNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yaotang Deng
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Yuxia Yan
- Department of Biostatistics, School of Public HealthSouthern Medical UniversityGuangzhouChina
| | - Matthias P. Mayer
- Center for Molecular Biology of Heidelberg University (ZMBH)DKFZ‐ZMBH‐AllianceHeidelbergGermany
| | - Xuemei Chen
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina,Center for Molecular Biology of Heidelberg University (ZMBH)DKFZ‐ZMBH‐AllianceHeidelbergGermany
| | - Fei Zou
- Department of Occupational Health and MedicineGuangdong Provincial Key Laboratory of Tropical Disease ResearchSchool of Public HealthSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
75
|
Sharifi M, Hosseinali SH, Saboury AA, Szegezdi E, Falahati M. Involvement of planned cell death of necroptosis in cancer treatment by nanomaterials: Recent advances and future perspectives. J Control Release 2019; 299:121-137. [DOI: 10.1016/j.jconrel.2019.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/31/2022]
|
76
|
You Y, Wang R, Shao N, Zhi F, Yang Y. Luteolin suppresses tumor proliferation through inducing apoptosis and autophagy via MAPK activation in glioma. Onco Targets Ther 2019; 12:2383-2396. [PMID: 30992674 PMCID: PMC6445239 DOI: 10.2147/ott.s191158] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Glioma is a malignant tumor that originates in the brain and spine and is difficult to be completely removed. Though glioma patients receive active treatment, the survival rate is still poor. Therefore, it is urgent to discover a new medicine to treat glioma patients in order to improve the survival rate. In this study, we explored the anticancer effect and the potential mechanism of luteolin on glioma in vitro. MATERIALS AND METHODS Cell viability was determined by Cell Counting Kit-8 (CCK-8) assay. Fluorescent microscopy and flow cytometry analysis were used to determine the cellular apoptosis. Western blot analysis was performed to explore the changes in protein expression. Quantitative reverse transcription-PCR (qRT-PCR) analysis was utilized to evaluate the expression level of the tumor suppressor miR-124-3p. RESULTS CCK-8 assays indicated that luteolin significantly inhibited glioma cell proliferation in a time- and dose-dependent manner. Fluorescent microscopy and flow cytometry analysis confirmed that luteolin induced glioma cell apoptosis. Western blot analysis showed that luteolin induced cellular apoptosis in glioma cells via MAPK activation (JNK, ERK, and p38). Luteolin stimulated the death receptor (FADD) to regulate the apoptosis proteins (Caspase-8, Caspase-3, and PARP). Luteolin increased the expression levels of LC3B II/I and downregulated the level of p62 that promotes cell autophagy. Finally, qRT-PCR confirmed that luteolin upregulated the expression levels of miR-124-3p. CONCLUSION These findings illustrate that luteolin may be a potential drug for glioma treatment.
Collapse
Affiliation(s)
- Yijie You
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China,
| | - Rong Wang
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
| | - Naiyuan Shao
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China,
| | - Feng Zhi
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
| | - Yilin Yang
- Department of Neurosurgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China, ,
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, China,
| |
Collapse
|
77
|
Kriel J, Loos B. The good, the bad and the autophagosome: exploring unanswered questions of autophagy-dependent cell death. Cell Death Differ 2019; 26:640-652. [PMID: 30659234 PMCID: PMC6460391 DOI: 10.1038/s41418-018-0267-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/04/2018] [Accepted: 12/07/2018] [Indexed: 01/15/2023] Open
Abstract
The recent discovery of autosis as a variant of autophagy-dependent cell death has challenged the conventional understanding of cell death and programmed cell death in cellular decision making. In contrast to previous accounts of distinct cell death modalities, autosis occurs with high autophagic activity, in the absence of apoptotic and necrotic markers and yet is not fully regulated by typical autophagy markers. Given the metabolic importance of autophagic responses and the extensive cross-talk with both apoptosis and necrosis signalling, the classical and morphotype-driven characterization of cell death as pre-determined subroutines is being increasingly called into question. Furthermore, the conflicting evidence with regards to cell death induction through autophagy modulation in various cancer models highlights the lack of consensus over the extent to which autophagy assists in cell death ontrol and whether it is capable of being a bona fide lethal process. This review evaluates the evidence and context of autophagy-dependent cell death and delineates the role of an autophagic flux threshold associated with 'lethal' and 'non-lethal' autophagy and its role in autosis control. In doing so, cancer treatment avenues will be explored with regards to precision modulation of tumour autophagic flux to ascertain whether autosis induction may present a novel therapeutic strategy.
Collapse
Affiliation(s)
- Jurgen Kriel
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, 7600, South Africa
| | - Ben Loos
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, 7600, South Africa.
| |
Collapse
|
78
|
Jaber FA, Khan NM, Ansari MY, Al-Adlaan AA, Hussein NJ, Safadi FF. Autophagy plays an essential role in bone homeostasis. J Cell Physiol 2019; 234:12105-12115. [PMID: 30820954 DOI: 10.1002/jcp.27071] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/29/2018] [Indexed: 12/20/2022]
Abstract
Autophagy is very critical for multiple cellular processes. Autophagy plays a critical role in bone cell differentiation and function.
Collapse
Affiliation(s)
- Fatima A Jaber
- Department of Biology, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazir M Khan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Mohammad Y Ansari
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio
| | - Asaad A Al-Adlaan
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Nazar J Hussein
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio
| | - Fayez F Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED) School of Medicine, Rootstown, Ohio.,School of Biomedical Sciences, Kent State University, Kent, Ohio.,Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, Ohio.,Department of Orthopedic Surgery, SUMMA Health System, Akron, Ohio.,Rebecca D. Considine Research Institute Akron Children's Hospital, Akron, Ohio
| |
Collapse
|
79
|
LncRNA H19 overexpression induces bortezomib resistance in multiple myeloma by targeting MCL-1 via miR-29b-3p. Cell Death Dis 2019; 10:106. [PMID: 30728351 PMCID: PMC6365572 DOI: 10.1038/s41419-018-1219-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
Abstract
Radiotherapy, chemotherapy, autologous/allogeneic stem cell transplantation, and targeted drug therapy are currently available therapeutic options for multiple myeloma (MM), but the clinical outcome remains unsatisfactory owing to frequent occurrence of drug resistance. Anti apoptosis is one of the main mechanisms to mediate drug resistance. Studies have shown that MCL-1 plays a key role in the growth of cancer cells “escaping” drug attacks, but the underlying mechanism remains unclear. Our previous study demonstrated that lncRNA H19 was highly expressed in the serum of MM patients. Bioinformatics predicts that miR-29b-3p is the downstream target gene, and MCL-1 is the downstream target protein of miR-29b-3p. Therefore, we speculated that MCL-1 may be involved in the occurrence of drug resistance through epigenetics. On the basis of these previous findings, the present study was intended to explore the biological function of H19, interactions between the downstream target genes, and the effect of H19 on BTZ resistance of myeloma cells. In addition, in vivo experiments we have also confirmed that H19 promoted tumor growth and may develop resistance to bortezomib partly. It was found that H19 reduced cell sensitivity to the chemotherapeutic drug BTZ by working as a miRNA sponge to inhibit the expression of miR-29b-3p, enhance MCL-1 transcriptional translation and inhibit apoptosis. These findings may help gain insights into the molecular mechanism of acquired BTZ resistance and develop new drug targets for the clinical treatment of MM.
Collapse
|
80
|
Therapeutic Modulation of Autophagy in Leukaemia and Lymphoma. Cells 2019; 8:cells8020103. [PMID: 30704144 PMCID: PMC6406467 DOI: 10.3390/cells8020103] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/26/2019] [Accepted: 01/28/2019] [Indexed: 02/07/2023] Open
Abstract
Haematopoiesis is a tightly orchestrated process where a pool of hematopoietic stem and progenitor cells (HSPCs) with high self-renewal potential can give rise to both lymphoid and myeloid lineages. The HSPCs pool is reduced with ageing resulting in few HSPC clones maintaining haematopoiesis thereby reducing blood cell diversity, a phenomenon called clonal haematopoiesis. Clonal expansion of HSPCs carrying specific genetic mutations leads to increased risk for haematological malignancies. Therefore, it comes as no surprise that hematopoietic tumours develop in higher frequency in elderly people. Unfortunately, elderly patients with leukaemia or lymphoma still have an unsatisfactory prognosis compared to younger ones highlighting the need to develop more efficient therapies for this group of patients. Growing evidence indicates that macroautophagy (hereafter referred to as autophagy) is essential for health and longevity. This review is focusing on the role of autophagy in normal haematopoiesis as well as in leukaemia and lymphoma development. Attenuated autophagy may support early hematopoietic neoplasia whereas activation of autophagy in later stages of tumour development and in response to a variety of therapies rather triggers a pro-tumoral response. Novel insights into the role of autophagy in haematopoiesis will be discussed in light of designing new autophagy modulating therapies in hematopoietic cancers.
Collapse
|
81
|
Xu J, Su Y, Xu A, Fan F, Mu S, Chen L, Chu Z, Zhang B, Huang H, Zhang J, Deng J, Ai L, Sun C, Hu Y. miR-221/222-Mediated Inhibition of Autophagy Promotes Dexamethasone Resistance in Multiple Myeloma. Mol Ther 2019; 27:559-570. [PMID: 30765325 DOI: 10.1016/j.ymthe.2019.01.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/26/2022] Open
Abstract
Inherent or acquired resistance to chemotherapeutic drugs is still an obstacle for the treatment of multiple myeloma (MM). MicroRNA dysregulation is related to the development of chemoresistance in cancers. However, its role in chemoresistance of MM is largely unknown. Here we demonstrated that miR-221/222 were upregulated in plasma cells from patients with MM, especially those with relapsed or refractory disease. Moreover, expression levels of miR-221/222 were inversely correlated with dexamethasone (Dex) sensitivity of human MM cell lines. Importantly, we found that Dex induced pro-death autophagy in MM cells and the inhibition of autophagy significantly decreased Dex-induced cell death. Mechanistically, autophagy-related gene 12 (ATG12) was identified as a novel target gene of miR-221/222, and miR-221/222 overexpression inhibited autophagy by directly targeting ATG12 and the p27kip (p27)-mammalian target of rapamycin (mTOR) pathway. Indeed, Dex treatment decreased the expression of miR-221/222, thereby activating the ATG12/p27-mTOR autophagy-regulatory axis and inducing cell death in Dex-sensitive MM cells. Furthermore, both in vitro and in vivo results showed that the inhibitions of miR-221/222 increased the expression of ATG12 and p27 and functionally induced extended autophagy and cell death of MM cells. In conclusion, our findings demonstrated the crucial role of the miR-221/222-ATG12/p27-mTOR autophagy-regulatory axis in Dex resistance of MM, and they suggest potential prediction and treatment strategies for glucocorticoid resistance.
Collapse
Affiliation(s)
- Jian Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yan Su
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fengjuan Fan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shidai Mu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangbo Chu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haifan Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiasi Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
82
|
Torres-López L, Maycotte P, Liñán-Rico A, Liñán-Rico L, Donis-Maturano L, Delgado-Enciso I, Meza-Robles C, Vásquez-Jiménez C, Hernández-Cruz A, Dobrovinskaya O. Tamoxifen induces toxicity, causes autophagy, and partially reverses dexamethasone resistance in Jurkat T cells. J Leukoc Biol 2019; 105:983-998. [PMID: 30645008 DOI: 10.1002/jlb.2vma0818-328r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 01/29/2023] Open
Abstract
Estrogens demonstrate biological activity in numerous organ systems, including the immune system, and exert their effects through estrogen receptors (ER) of two types: intracellular ERα and ERβ that activate transcriptional factors and membrane G protein-coupled ER GPER. The latter is capable to mediate fast activation of cytosolic signaling pathways, influencing transcriptional events in response to estrogens. Tamoxifen (TAM), widely used in chemotherapy of ERα-positive breast cancer, is considered as an ERα antagonist and GPER agonist. TAM was shown to possess "off-target" cytotoxicity, not related to ER in various tumor types. The present work was designed to study biological effects of TAM on the glucocorticoid (GC)-resistant cell line Jurkat, derived from acute lymphoblastic leukemia of T lineage (T-ALL). We have shown that T-ALL cell lines, in contrast to healthy T cells, express only GPER, but not ERα or ERβ. TAM compromised mitochondrial function and reduced the viability and proliferation of Jurkat cells. Additionally, TAM induced autophagy in a GPER-dependent manner. Gene expression profiling revealed the up-regulation of autophagy-related gene ATG5. Interestingly, TAM sensitized Jurkat cells to dexamethasone (DEX) treatment, which may be related to its capacity to cause autophagy. We suggest that TAM-based adjuvant therapy may represent a novel strategy in T-ALL patients handling.
Collapse
Affiliation(s)
- Liliana Torres-López
- University Center for Biomedical Research, University of Colima, Colima, Mexico.,Faculty for Chemical Sciences, University of Colima, Colima, Mexico
| | - Paola Maycotte
- CONACYT-Biomedical Research Center of the East, Mexican Social Security Institute, Puebla, Mexico
| | - Andrómeda Liñán-Rico
- CONACYT-University Center for Biomedical Research, University of Colima, Colima, Mexico
| | - Liliana Liñán-Rico
- University Center for Biomedical Research, University of Colima, Colima, Mexico
| | - Luis Donis-Maturano
- Ensenada Biomedical Innovation Department, Center for Scientific Research and Higher Education, Ensenada, Baja California, Mexico
| | - Iván Delgado-Enciso
- Medical School, University of Colima and Cancerology Institute of Colima State, Health Services, Colima, Mexico
| | - Carmen Meza-Robles
- Medical School, University of Colima and Cancerology Institute of Colima State, Health Services, Colima, Mexico
| | | | - Arturo Hernández-Cruz
- National Laboratory of Channelopathies (LaNCa), National Autonomous University of Mexico, Mexico City, Mexico.,Department of Cognitive Neuroscience-Institute of Cellular Physiology, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oxana Dobrovinskaya
- University Center for Biomedical Research, University of Colima, Colima, Mexico
| |
Collapse
|
83
|
Qin C, Zhang R, Lang Y, Shao A, Xu A, Feng W, Han J, Wang M, He W, Yu C, Tang J. Bclaf1 critically regulates the type I interferon response and is degraded by alphaherpesvirus US3. PLoS Pathog 2019; 15:e1007559. [PMID: 30682178 PMCID: PMC6364948 DOI: 10.1371/journal.ppat.1007559] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/06/2019] [Accepted: 01/03/2019] [Indexed: 01/12/2023] Open
Abstract
Type I interferon response plays a prominent role against viral infection, which is frequently disrupted by viruses. Here, we report Bcl-2 associated transcription factor 1 (Bclaf1) is degraded during the alphaherpesvirus Pseudorabies virus (PRV) and Herpes simplex virus type 1 (HSV-1) infections through the viral protein US3. We further reveal that Bclaf1 functions critically in type I interferon signaling. Knockdown or knockout of Bclaf1 in cells significantly impairs interferon-α (IFNα) -mediated gene transcription and viral inhibition against US3 deficient PRV and HSV-1. Mechanistically, Bclaf1 maintains a mechanism allowing STAT1 and STAT2 to be efficiently phosphorylated in response to IFNα, and more importantly, facilitates IFN-stimulated gene factor 3 (ISGF3) binding with IFN-stimulated response elements (ISRE) for efficient gene transcription by directly interacting with ISRE and STAT2. Our studies establish the importance of Bclaf1 in IFNα-induced antiviral immunity and in the control of viral infections.
Collapse
Affiliation(s)
- Chao Qin
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Rui Zhang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yue Lang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Anwen Shao
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Aotian Xu
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wenhai Feng
- Department of Microbiology and Immunology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jun Han
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengdong Wang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wanwei He
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Cuilian Yu
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Tang
- State Key Laboratory of Agrobiotechnology and College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
84
|
Denton D, Kumar S. Autophagy-dependent cell death. Cell Death Differ 2018; 26:605-616. [PMID: 30568239 DOI: 10.1038/s41418-018-0252-y] [Citation(s) in RCA: 547] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 11/09/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Autophagy-dependent cell death can be defined as cell demise that has a strict requirement of autophagy. Although autophagy often accompanies cell death following many toxic insults, the requirement of autophagic machinery for cell death execution, as established through specific genetic or chemical inhibition of the process, is highly contextual. During animal development, perhaps the best validated model of autophagy-dependent cell death is the degradation of the larval midgut during larval-pupal metamorphosis, where a number of key autophagy genes are required for the removal of the tissues. Surprisingly though, even in the midgut, not all of the 'canonical' autophagic machinery appears to be required. In other organisms and cancer cells many variations of autophagy-dependent cell death are apparent, pointing to the lack of a unifying cell death pathway. It is thus possible that components of the autophagy machinery are selectively utilised or repurposed for this type of cell death. In this review, we discuss examples of cell death that utilise autophagy machinery (or part thereof), the current knowledge of the complexity of autophagy-dependent cellular demise and the potential mechanisms and regulatory pathways involved in such cell death.
Collapse
Affiliation(s)
- Donna Denton
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, SA, 5001, Australia.
| |
Collapse
|
85
|
Nazim U, Park S. Luteolin sensitizes human liver cancer cells to TRAIL‑induced apoptosis via autophagy and JNK‑mediated death receptor 5 upregulation. Int J Oncol 2018; 54:665-672. [DOI: 10.3892/ijo.2018.4633] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/19/2018] [Indexed: 11/05/2022] Open
Affiliation(s)
- Uddin Nazim
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Sang‑Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| |
Collapse
|
86
|
Akkoç Y, Gözüaçık D. Autophagy and liver cancer. TURKISH JOURNAL OF GASTROENTEROLOGY 2018; 29:270-282. [PMID: 29755011 DOI: 10.5152/tjg.2018.150318] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Autophagy is a key biological phenomenon conserved from yeast to mammals. Under basal conditions, activation of autophagy leads to the protein degradation as well as damaged organelles for maintaining cellular homeostasis. Deregulation of autophagy has been identified as a key mechanism contributing to the pathogenesis and progression of several liver diseases, including hepatocellular carcinoma (HCC), one of the most common and mortal types of cancer. Currently used treatment strategies in patients with HCC result in variable success rates. Therefore, novel early diagnosis and treatment techniques should be developed. Manipulation of autophagy may improve responses of cancer cell to treatments and provide novel targeted therapy options for HCC. In this review, we summarized how our understanding of autophagy-cell death connection may have an impact on HCC therapy.
Collapse
Affiliation(s)
- Yunus Akkoç
- Department of Molecular Biology, Genetics and Bioengineering, Sabancı University School of Engineering and Natural Sciences, İstanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, İstanbul, Turkey
| | - Devrim Gözüaçık
- Department of Molecular Biology, Genetics and Bioengineering, Sabancı University School of Engineering and Natural Sciences, İstanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, İstanbul, Turkey
| |
Collapse
|
87
|
Ge L, Wang Q, Hu S, Yang X. Rs217727 polymorphism in H19 promotes cell apoptosis by regulating the expressions of H19 and the activation of its downstream signaling pathway. J Cell Physiol 2018; 234:7279-7291. [PMID: 30362559 DOI: 10.1002/jcp.27485] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/06/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND The objective of the current study was to explore the role of H19 rs217727 polymorphism in the control of hepatocellular carcinoma (HCC). METHOD The Student's t test, Cox regression, and Kaplan-Meier analyses were used to clarify whether the H19 rs217727 polymorphism played an important role in the development of HCC. Real-time polymerase chain reaction (PCR) and western-blot analysis were carried out to measure the levels of H19, microRNA (miR)-675, FAS-associated death domain (FADD), caspase-8, and caspase-3 among H19 CC, CT, and TT groups, as well as in cells transfected with H19/si-H19, or miR-675 mimic/inhibitor. The MTT assay, colony formation assay, and flow cytometry assay were performed to detect the effect of H19/miR-675 on cell viability, cell colony formation, and cell apoptosis. RESULT T allele of H19 rs217727 polymorphism apparently increased the survival rate of patients with HCC. Meanwhile, H19 enhanced miR-675 expression but reduced the mRNA and protein levels of FADD, caspase-3, and caspase-8. The T allele of H19 rs217727 polymorphism apparently increased the apoptotic rate of HCC cells. Furthermore, FADD was a virtual target gene of miR-675 with a potential "hit" located in the 3'-untranslated region (UTR) of FADD, whereas H19 inhibited FADD expression via increasing the expression of miR-675. Moreover, H19 upregulated the expression of miR-675 whereas reducing the expression of FADD, caspase-3, and caspase-8. Finally, H19 and miR-675 promoted cell proliferation and cell colony formation but repressed cell apoptosis. CONCLUSION In summary, the above findings demonstrated that the polymorphism of rs217727 in H19 was associated with HCC via the H19/miR-675/FADD/caspase-8/caspase-3/apoptosis signaling pathway.
Collapse
Affiliation(s)
- Lili Ge
- Henan Provincial Key Labratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University (Henan Children's Hospital, Zhengzhou Children's Hospital), Zhengzhou, Henan, China
| | - Qinglei Wang
- Department of Pediatric Orthopedics, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan, China
| | - Shengnan Hu
- Department of Liver Disease, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoang Yang
- Department of Liver Disease, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
88
|
Bialik S, Dasari SK, Kimchi A. Autophagy-dependent cell death – where, how and why a cell eats itself to death. J Cell Sci 2018; 131:131/18/jcs215152. [DOI: 10.1242/jcs.215152] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT
Autophagy as a means of cell killing was first advanced by Clark's phenotypic description of ‘Type II autophagic cell death’ in 1990. However, this phenomenon later came into question, because the presence of autophagosomes in dying cells does not necessarily signify that autophagy is the cause of demise, but rather may reflect the efforts of the cell to prevent it. Resolution of this issue comes from a more careful definition of autophagy-dependent cell death (ADCD) as a regulated cell death that is shown experimentally to require different components of the autophagy machinery without involvement of alternative cell death pathways. Following these strict criteria, ADCD has been validated in both lower model organisms and mammalian cells, highlighting its importance for developmental and pathophysiological cell death. Recently, researchers have defined additional morphological criteria that characterize ADCD and begun to explore how the established, well-studied autophagy pathway is subverted from a survival to a death function. This Review explores validated models of ADCD and focuses on the current understanding of the mechanisms by which autophagy can kill a cell.
Collapse
Affiliation(s)
- Shani Bialik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Santosh K. Dasari
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
89
|
Desantis V, Saltarella I, Lamanuzzi A, Mariggiò MA, Racanelli V, Vacca A, Frassanito MA. Autophagy: A New Mechanism of Prosurvival and Drug Resistance in Multiple Myeloma. Transl Oncol 2018; 11:1350-1357. [PMID: 30196237 PMCID: PMC6132177 DOI: 10.1016/j.tranon.2018.08.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 12/25/2022] Open
Abstract
Autophagy is an intracellular self-degradative process that balances cell energy source and regulates tissue homeostasis. In physiological condition, autophagy funnels cytoplasmic constituents to autophagolysosomes for degradation and is an alternative way for cell-death behavior. Here, we inspected autophagy as a prosurvival mechanism essential for drug resistance in multiple myeloma (MM). Accordingly, autophagy inhibitors used in association to conventional anti-MM drugs might enforce the effect against resistant MM plasma cells and render autophagy a new therapeutic target.
Collapse
Affiliation(s)
- V Desantis
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - I Saltarella
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - A Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - M A Mariggiò
- Department of Biomedical Sciences and Human Oncology, Unit of General Pathology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - V Racanelli
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy.
| | - M A Frassanito
- Department of Biomedical Sciences and Human Oncology, Unit of General Pathology, University of Bari Aldo Moro Medical School, Bari, Italy
| |
Collapse
|
90
|
Gudipaty SA, Conner CM, Rosenblatt J, Montell DJ. Unconventional Ways to Live and Die: Cell Death and Survival in Development, Homeostasis, and Disease. Annu Rev Cell Dev Biol 2018; 34:311-332. [PMID: 30089222 DOI: 10.1146/annurev-cellbio-100616-060748] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Balancing cell death and survival is essential for normal development and homeostasis and for preventing diseases, especially cancer. Conventional cell death pathways include apoptosis, a form of programmed cell death controlled by a well-defined biochemical pathway, and necrosis, the lysis of acutely injured cells. New types of regulated cell death include necroptosis, pyroptosis, ferroptosis, phagoptosis, and entosis. Autophagy can promote survival or can cause death. Newly described processes of anastasis and resuscitation show that, remarkably, cells can recover from the brink of apoptosis or necroptosis. Important new work shows that epithelia achieve homeostasis by extruding excess cells, which then die by anoikis due to loss of survival signals. This mechanically regulated process both maintains barrier function as cells die and matches rates of proliferation and death. In this review, we describe these unconventional ways in which cells have evolved to die or survive, as well as the contributions that these processes make to homeostasis and cancer.
Collapse
Affiliation(s)
- Swapna A Gudipaty
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christopher M Conner
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| | - Jody Rosenblatt
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| |
Collapse
|
91
|
Lu Y, Wang Y, Xu H, Shi C, Jin F, Li W. Profilin 1 induces drug resistance through Beclin1 complex-mediated autophagy in multiple myeloma. Cancer Sci 2018; 109:2706-2716. [PMID: 29945297 PMCID: PMC6125445 DOI: 10.1111/cas.13711] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/19/2018] [Indexed: 01/01/2023] Open
Abstract
Autophagy plays an important role in multiple myeloma (MM) for homeostasis, survival and drug resistance, but which genes participate in this process is unclear. We identified several cytoskeleton genes upregulated in MM patients by gene expression profiling (GEP) datasets; in particular, patients with high profilin 1 (PFN1) expression had poor prognosis in MM. In vitro, overexpressed PFN1 promotes proliferation and bortezomib (BTZ) resistance in MM cells. Further study indicated overexpression of PFN1 significantly promoted the process of autophagy and induced BTZ resistance in MM. Otherwise, knockdown of PFN1 blocked autophagy and sensitized MM to BTZ. Co‐immunoprecipitation in MM cells indicated that PFN1 could bind Beclin1 complex and promote the initiation of autophagy. Inhibition of autophagy by blocking the formation of Beclin1 complex could reverse the phenotype of BTZ resistance in MM. Our findings suggested that PFN1 could promote autophagy through taking part in Beclin1 complex and contribute to BTZ resistance, which may become a novel molecular target in the therapy of MM.
Collapse
Affiliation(s)
- Yichen Lu
- Cancer Center, the First Bethune Hospital of Jilin University, Jilin, China
| | - Ya Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), National Key Clinical Specialty, Xiangya Hospital, Central South University, Hunan, China
| | - He Xu
- Cancer Center, the First Bethune Hospital of Jilin University, Jilin, China
| | - Chen Shi
- Cancer Center, the First Bethune Hospital of Jilin University, Jilin, China
| | - Fengyan Jin
- Cancer Center, the First Bethune Hospital of Jilin University, Jilin, China
| | - Wei Li
- Cancer Center, the First Bethune Hospital of Jilin University, Jilin, China
| |
Collapse
|
92
|
Zhu J, Cai Y, Xu K, Ren X, Sun J, Lu S, Chen J, Xu P. Beclin1 overexpression suppresses tumor cell proliferation and survival via an autophagy‑dependent pathway in human synovial sarcoma cells. Oncol Rep 2018; 40:1927-1936. [PMID: 30066884 PMCID: PMC6111547 DOI: 10.3892/or.2018.6599] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Beclin1 is an important autophagy‑related prot-ein, which is involved in both autophagy and apoptosis. In recent years, the antitumor effect of Beclin1 has received increased attention. In the present study, we established a stable Beclin1‑overexpressing cell line with SW982 human synovial sarcoma cells. We found that Beclin1 overexpression decreased the cell viability, inhibited proliferation and induced apoptosis in SW982 cells. The expression levels of Bcl‑2 and PCNA were decreased, while the levels of cleaved‑caspase‑3 and cleaved‑PARP were increased. Beclin1 is closely related with autophagy, thus the autophagy‑related markers LC3 and p62 were detected by western blot analysis, and transmission electron microscopy was used to observe autophagosomes. The results showed that the expression level of LC3II was increased and that of p62 was decreased. Moreover, many double membrane‑enclosed autophagosomes were found in cells with Beclin1 overexpression, which indicated that the autophagic activity was enhanced. To explore the effect of autophagy on the viability of SW982 cells, Atg5 was knocked down using siRNA to inhibit the autophagic activity. We found that autophagy contributed to the decrease in cell viability. Knockdown of Atg5 increased the viability and decreased the apoptotic rate of SW982 cells with Beclin1 overexpression. The expression level of Bcl‑2 was increased, while the expression levels of cleaved‑caspase‑3 and cleaved‑PARP were decreased. We also found that the Akt/Bcl‑2/caspase‑9 pathway was involved. The phosphorylation of AKT was positively correlated with cell viability. The cleavage of caspase‑9 was increased by Beclin1 overexpression and decreased by inhibition of autophagy. Altogether, our results suggested that both autophagy and apoptosis contributed to the antitumor effect of Beclin1 in SW982 cells.
Collapse
Affiliation(s)
- Jialin Zhu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoyu Ren
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Jian Sun
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shemin Lu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Jinghong Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Institute of Endemic Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
93
|
He L, Zhang J, Zhao J, Ma N, Kim SW, Qiao S, Ma X. Autophagy: The Last Defense against Cellular Nutritional Stress. Adv Nutr 2018; 9:493-504. [PMID: 30032222 PMCID: PMC6054220 DOI: 10.1093/advances/nmy011] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Homeostasis of nutrient metabolism is critical for maintenance of the normal physiologic status of the cell and the integral health of humans and mammals. In vivo, there is a highly efficient and precise process involved in nutrient recycling and organelle cleaning. This process is named autophagy, and it can be induced in response to the dynamic change of nutrients. When cells face nutritional stress, such as stress caused by nutrient deficiency or nutrient excess, the autophagy pathway will be activated. Generally, when nutrients are withdrawn, cells will sense the signs of starvation and respond. AMP-activated protein kinase and the mammalian target of rapamycin, two of the major metabolic kinases, are responsible for monitoring cellular energy and the concentration of amino acids, respectively. Nutrient excess also induces autophagy, mainly via the reactive oxygen species and endoplasmic reticulum stress pathway. When nutritional stress activates the autophagy pathway, the nutrients or damaged organelles will be recycled for cell survival. However, if autophagy is overwhelmingly induced, autophagic cell death will possibly occur. The balance of the autophagy induction is the crucial factor for cell survival or death. Herein, we summarize the current knowledge on the induction of autophagy, the autophagy response under nutritional stresses, and autophagic cell death and related diseases, which will highlight the process of nutritional stress-induced autophagy and its important physiologic and/or pathologic roles in cell metabolism and diseases, and shed light on the research into the mechanism and clinical applications of autophagy induced by nutritional stresses.
Collapse
Affiliation(s)
- Long He
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, China
| | - Jie Zhang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, China
| | - Jinshan Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China
| | - Ning Ma
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, China
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing, China,College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, China,Department of Internal Medicine, Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Address correspondence to XM (e-mail: )
| |
Collapse
|
94
|
Humphreys L, Espona-Fiedler M, Longley DB. FLIP as a therapeutic target in cancer. FEBS J 2018; 285:4104-4123. [PMID: 29806737 DOI: 10.1111/febs.14523] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
One of the classic hallmarks of cancer is disruption of cell death signalling. Inhibition of cell death promotes tumour growth and metastasis, causes resistance to chemo- and radiotherapies as well as targeted agents, and is frequently due to overexpression of antiapoptotic proteins rather than loss of pro-apoptotic effectors. FLIP is a major apoptosis-regulatory protein frequently overexpressed in solid and haematological cancers, in which its high expression is often correlated with poor prognosis. FLIP, which is expressed as long (FLIP(L)) and short (FLIP(S)) splice forms, achieves its cell death regulatory functions by binding to FADD, a critical adaptor protein which links FLIP to the apical caspase in the extrinsic apoptotic pathway, caspase-8, in a number of cell death regulating complexes, such as the death-inducing signalling complexes (DISCs) formed by death receptors. FLIP also plays a key role (together with caspase-8) in regulating another form of cell death termed programmed necrosis or 'necroptosis', as well as in other key cellular processes that impact cell survival, including autophagy. In addition, FLIP impacts activation of the intrinsic mitochondrial-mediated apoptotic pathway by regulating caspase-8-mediated activation of the pro-apoptotic Bcl-2 family member Bid. It has been demonstrated that FLIP can not only inhibit death receptor-mediated apoptosis, but also cell death induced by a range of clinically relevant chemotherapeutic and targeted agents as well as ionizing radiation. More recently, key roles for FLIP in promoting the survival of immunosuppressive tumour-promoting immune cells have been discovered. Thus, FLIP is of significant interest as an anticancer therapeutic target. In this article, we review FLIP's biology and potential ways of targeting this important tumour and immune cell death regulator.
Collapse
Affiliation(s)
- Luke Humphreys
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Margarita Espona-Fiedler
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Drug Resistance Group, Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
95
|
Duarte AI, Sjögren M, Santos MS, Oliveira CR, Moreira PI, Björkqvist M. Dual Therapy with Liraglutide and Ghrelin Promotes Brain and Peripheral Energy Metabolism in the R6/2 Mouse Model of Huntington's Disease. Sci Rep 2018; 8:8961. [PMID: 29895889 PMCID: PMC5997749 DOI: 10.1038/s41598-018-27121-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Neuronal loss alongside altered energy metabolism, are key features of Huntington’s disease (HD) pathology. The orexigenic gut-peptide hormone ghrelin is known to stimulate appetite and affect whole body energy metabolism. Liraglutide is an efficient anti-type 2 diabetes incretin drug, with neuroprotective effects alongside anorectic properties. Combining liraglutide with the orexigenic peptide ghrelin may potentially promote brain/cognitive function in HD. The R6/2 mouse model of HD exhibits progressive central pathology, weight loss, deranged glucose metabolism, skeletal muscle atrophy and altered body composition. In this study, we targeted energy metabolism in R6/2 mice using a co-administration of liraglutide and ghrelin. We investigated their effect on brain cortical hormone-mediated intracellular signalling pathways, metabolic and apoptotic markers, and the impact on motor function in HD. We here demonstrate that liraglutide, alone or together with ghrelin (subcutaneous daily injections of 150 µg/kg (ghrelin) and 0.2 mg/kg (liraglutide), for 2 weeks), normalized glucose homeostatic features in the R6/2 mouse, without substantially affecting body weight or body composition. Liraglutide alone decreased brain cortical active GLP-1 and IGF-1 levels in R6/2 mice, alongside higher ADP levels. Liraglutide plus ghrelin decreased brain insulin, lactate, AMP and cholesterol levels in R6/2 mice. Taken together, our findings encourage further studies targeting energy metabolism in HD.
Collapse
Affiliation(s)
- Ana I Duarte
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal. .,Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.
| | - Marie Sjögren
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Maria S Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Life Sciences Department, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - Catarina R Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Biochemistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Björkqvist
- Brain Disease Biomarker Unit, Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
96
|
Manni S, Carrino M, Semenzato G, Piazza F. Old and Young Actors Playing Novel Roles in the Drama of Multiple Myeloma Bone Marrow Microenvironment Dependent Drug Resistance. Int J Mol Sci 2018; 19:ijms19051512. [PMID: 29783691 PMCID: PMC5983700 DOI: 10.3390/ijms19051512] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is the second most frequent hematologic cancer. In addition to the deleterious effects of neoplastic plasma cell growth and spreading during the disease evolution, this tumor is characterized by the serious pathological consequences due to the massive secretion of monoclonal immunoglobulins and by the derangement of bone physiology with progressive weakening of the skeleton. Despite significant progresses having been made in the last two decades in the therapeutic management of this plasma cell tumor, MM remains invariably lethal, due to its extremely complex genetic architecture and to the constant protection it receives from the tumor niche, which is represented by the bone marrow microenvironment. While it is predictable that the discovery of novel therapies against the first of these two pathobiological features will take a longer time, the identification of the cellular and molecular mechanisms underlying the pro-growth effects of the myeloma milieu is a task that could lead to the development of novel treatments in a shorter timeframe. In this regard, aside from known “old” determinants of the cross-talk between bone marrow and MM cells, “young” cellular and molecular factors are now emerging, taking the scene of this complex neoplastic setting. In this review we aimed at giving insights on the latest evidence of potentially-targetable modes that MM cells exploit to increase fitness and gain a survival advantage. The benefits coming from the derangements of stress-managing pathways, autophagy, transcriptional rewiring, and non-coding RNAs are examples of such methods that MM cells utilize to escape cell death, but that hopefully will offer novel targets for the ever-increasing anti-MM therapeutic armamentarium.
Collapse
Affiliation(s)
- Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Marilena Carrino
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Gianpietro Semenzato
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via N.Giustiniani 2, 35128 Padova, Italy.
- Venetian Institute of Molecular Medicine, Via G.Orus 2, 35129 Padova, Italy.
| |
Collapse
|
97
|
Cherian MA, Olson S, Sundaramoorthi H, Cates K, Cheng X, Harding J, Martens A, Challen GA, Tyagi M, Ratner L, Rauch D. An activating mutation of interferon regulatory factor 4 (IRF4) in adult T-cell leukemia. J Biol Chem 2018. [PMID: 29540473 DOI: 10.1074/jbc.ra117.000164] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The human T-cell leukemia virus-1 (HTLV-1) oncoprotein Tax drives cell proliferation and resistance to apoptosis early in the pathogenesis of adult T-cell leukemia (ATL). Subsequently, probably as a result of specific immunoediting, Tax expression is down-regulated and functionally replaced by somatic driver mutations of the host genome. Both amplification and point mutations of interferon regulatory factor 4 (IRF4) have been previously detected in ATL., K59R is the most common single-nucleotide variation of IRF4 and is found exclusively in ATL. High-throughput whole-exome sequencing revealed recurrent activating genetic alterations in the T-cell receptor, CD28, and NF-κB pathways. We found that IRF4, which is transcriptionally activated downstream of these pathways, is frequently mutated in ATL. IRF4 RNA, protein, and IRF4 transcriptional targets are uniformly elevated in HTLV-1-transformed cells and ATL cell lines, and IRF4 was bound to genomic regulatory DNA of many of these transcriptional targets in HTLV-1-transformed cell lines. We further noted that the K59R IRF4 mutant is expressed at higher levels in the nucleus than WT IRF4 and is transcriptionally more active. Expression of both WT and the K59R mutant of IRF4 from a constitutive promoter in retrovirally transduced murine bone marrow cells increased the abundance of T lymphocytes but not myeloid cells or B lymphocytes in mice. IRF4 may represent a therapeutic target in ATL because ATL cells select for a mutant of IRF4 with higher nuclear expression and transcriptional activity, and overexpression of IRF4 induces the expansion of T lymphocytes in vivo.
Collapse
Affiliation(s)
- Mathew A Cherian
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Sydney Olson
- the Department of Biology, University of Wisconsin, Madison, Wisconsin 53706, and
| | - Hemalatha Sundaramoorthi
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Kitra Cates
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Xiaogang Cheng
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - John Harding
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Andrew Martens
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Grant A Challen
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Manoj Tyagi
- the Computational Biology Branch, National Center for Biotechnology Information, National Institutes of Health, Bethesda, Maryland 20892
| | - Lee Ratner
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110,
| | - Daniel Rauch
- From the Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
98
|
Vegliante R, Ciriolo MR. Autophagy and Autophagic Cell Death: Uncovering New Mechanisms Whereby Dehydroepiandrosterone Promotes Beneficial Effects on Human Health. VITAMINS AND HORMONES 2018; 108:273-307. [PMID: 30029730 DOI: 10.1016/bs.vh.2018.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dehydroepiandrosterone (DHEA) is the most abundant steroid hormone in human serum and a precursor of sexual hormones. Its levels, which are maximum between the age of 20 and 30, dramatically decline with aging thus raising the question that many pathological conditions typical of the elderly might be associated with the decrement of circulating DHEA. Moreover, since its very early discovery, DHEA and its metabolites have been shown to be active in many pathophysiological contexts, including cardiovascular disease, brain disorders, and cancer. Indeed, treatment with DHEA has beneficial effects for the cure of these and many other pathologies in vitro, in vivo, and in patient studies. However, the molecular mechanisms underlying DHEA effects have been only partially elucidated. Autophagy is a self-digestive process, by which cell homeostasis is maintained, damaged organelles removed, and cell survival assured upon stress stimuli. However, high rate of autophagy is detrimental and leads to a form of programmed cell death known as autophagic cell death (ACD). In this chapter, we describe the process of autophagy and the morphological and biochemical features of ACD. Moreover, we analyze the beneficial effects of DHEA in several pathologies and the molecular mechanisms with particular emphasis on its regulation of cell death processes. Finally, we review data indicating DHEA and structurally related steroid hormones as modulators of both autophagy and ACD, a research field that opens new avenues in the therapeutic use of these compounds.
Collapse
Affiliation(s)
- Rolando Vegliante
- MN3T, The Microenvironmental Niche in Tumorigenesis and Targeted Therapy, Hopital Civil-Institut d'Hématologie et Immunologie, Strasbourg, France
| | - Maria R Ciriolo
- University of Rome 'Tor Vergata', Rome, Italy; IRCCS San Raffaele 'La Pisana', Rome, Italy.
| |
Collapse
|
99
|
Wang X, Wei X, Yuan Y, Sun Q, Zhan J, Zhang J, Tang Y, Li F, Ding L, Ye Q, Zhang H. Src-mediated phosphorylation converts FHL1 from tumor suppressor to tumor promoter. J Cell Biol 2018; 217:1335-1351. [PMID: 29434030 PMCID: PMC5881501 DOI: 10.1083/jcb.201708064] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 12/19/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023] Open
Abstract
FHL1 has been recognized for a long time as a tumor suppressor protein that associates with both the actin cytoskeleton and the transcriptional machinery. We present in this study a paradigm that phosphorylated FHL1 functions as an oncogenic protein by promoting tumor cell proliferation. The cytosolic tyrosine kinase Src interacts with and phosphorylates FHL1 at Y149 and Y272, which switches FHL1 from a tumor suppressor to a cell growth accelerator. Phosphorylated FHL1 translocates into the nucleus, where it binds to the transcription factor BCLAF1 and promotes tumor cell growth. Importantly, the phosphorylation of FHL1 is increased in tissues from lung adenocarcinoma patients despite the down-regulation of total FHL1 expression. Kindlin-2 was found to interact with FHL1 and recruit FHL1 to focal adhesions. Kindlin-2 competes with Src for binding to FHL1 and suppresses Src-mediated FHL1 phosphorylation. Collectively, we demonstrate that FHL1 can either suppress or promote tumor cell growth depending on the status of the sites for phosphorylation by Src.
Collapse
Affiliation(s)
- Xiang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Xiaofan Wei
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Yang Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Qingrui Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Jun Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Jing Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Yan Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Feng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| | - Lihua Ding
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Hongquan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Human Anatomy, Histology and Embryology, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, China
| |
Collapse
|
100
|
Horn S, Hughes MA, Schilling R, Sticht C, Tenev T, Ploesser M, Meier P, Sprick MR, MacFarlane M, Leverkus M. Caspase-10 Negatively Regulates Caspase-8-Mediated Cell Death, Switching the Response to CD95L in Favor of NF-κB Activation and Cell Survival. Cell Rep 2018; 19:785-797. [PMID: 28445729 PMCID: PMC5413585 DOI: 10.1016/j.celrep.2017.04.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/30/2016] [Accepted: 04/03/2017] [Indexed: 12/12/2022] Open
Abstract
Formation of the death-inducing signaling complex (DISC) initiates extrinsic apoptosis. Caspase-8 and its regulator cFLIP control death signaling by binding to death-receptor-bound FADD. By elucidating the function of the caspase-8 homolog, caspase-10, we discover that caspase-10 negatively regulates caspase-8-mediated cell death. Significantly, we reveal that caspase-10 reduces DISC association and activation of caspase-8. Furthermore, we extend our co-operative/hierarchical binding model of caspase-8/cFLIP and show that caspase-10 does not compete with caspase-8 for binding to FADD. Utilizing caspase-8-knockout cells, we demonstrate that caspase-8 is required upstream of both cFLIP and caspase-10 and that DISC formation critically depends on the scaffold function of caspase-8. We establish that caspase-10 rewires DISC signaling to NF-κB activation/cell survival and demonstrate that the catalytic activity of caspase-10, and caspase-8, is redundant in gene induction. Thus, our data are consistent with a model in which both caspase-10 and cFLIP coordinately regulate CD95L-mediated signaling for death or survival.
Collapse
Affiliation(s)
- Sebastian Horn
- Section of Molecular Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| | - Michelle A Hughes
- MRC Toxicology Unit, Hodgkin Building, PO Box 138, Lancaster Road, Leicester LE1 9HN, UK
| | - Ramon Schilling
- Section of Molecular Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Carsten Sticht
- Center for Medical Research, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Tencho Tenev
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Michaela Ploesser
- Section of Molecular Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Pascal Meier
- The Breakthrough Toby Robins Breast Cancer Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, Fulham Road, London SW3 6JB, UK
| | - Martin R Sprick
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Marion MacFarlane
- MRC Toxicology Unit, Hodgkin Building, PO Box 138, Lancaster Road, Leicester LE1 9HN, UK.
| | - Martin Leverkus
- Section of Molecular Dermatology, Department of Dermatology, Venereology, and Allergology, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany; Department of Dermatology and Allergology, Medical Faculty of the RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| |
Collapse
|