1
|
Kolahdouzmohammadi M, Pahlavan S, Sotoodehnejadnematalahi F, Tahamtani Y, Totonchi M. Activation of AMPK promotes cardiac differentiation by stimulating the autophagy pathway. J Cell Commun Signal 2023; 17:939-955. [PMID: 37040028 PMCID: PMC10409960 DOI: 10.1007/s12079-023-00744-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 03/22/2023] [Indexed: 04/12/2023] Open
Abstract
Autophagy, a critical catabolic process for cell survival against different types of stress, has a role in the differentiation of various cells, such as cardiomyocytes. Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is an energy-sensing protein kinase involved in the regulation of autophagy. In addition to its direct role in regulating autophagy, AMPK can also influence other cellular processes by regulating mitochondrial function, posttranslational acetylation, cardiomyocyte metabolism, mitochondrial autophagy, endoplasmic reticulum stress, and apoptosis. As AMPK is involved in the control of various cellular processes, it can influence the health and survival of cardiomyocytes. This study investigated the effects of an AMPK inducer (Metformin) and an autophagy inhibitor (Hydroxychloroquine) on the differentiation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). The results showed that autophagy was upregulated during cardiac differentiation. Furthermore, AMPK activation increased the expression of CM-specific markers in hPSC-CMs. Additionally, autophagy inhibition impaired cardiomyocyte differentiation by targeting autophagosome-lysosome fusion. These results indicate the significance of autophagy in cardiomyocyte differentiation. In conclusion, AMPK might be a promising target for the regulation of cardiomyocyte generation by in vitro differentiation of pluripotent stem cells.
Collapse
Affiliation(s)
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
2
|
Belyaeva E, Loginova N, Schroeder BA, Goldlust IS, Acharya A, Kumar S, Timashev P, Ulasov I. The spectrum of cell death in sarcoma. Biomed Pharmacother 2023; 162:114683. [PMID: 37031493 DOI: 10.1016/j.biopha.2023.114683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The balance between cell death and cell survival is a highly coordinated process by which cells break down and remove unnecessary or harmful materials in a controlled, highly regulated, and compartmentalized manner. Cell exposure to various stresses, such as oxygen starvation, a lack of nutrients, or exposure to radiation, can initiate autophagy. Autophagy is a carefully orchestrated process with multiple steps, each regulated by specific genes and proteins. Autophagy proteins impact cellular maintenance and cell fate in response to stress, and targeting this process is one of the most promising methods of anti-tumor therapy. It is currently not fully understood how autophagy affects different types of tumor cells, which makes it challenging to predict outcomes when this process is manipulated. In this review, we will explore the mechanisms of autophagy and investigate it as a potential and promising therapeutic target for aggressive sarcomas.
Collapse
Affiliation(s)
- Elizaveta Belyaeva
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Brett A Schroeder
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ian S Goldlust
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Arbind Acharya
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Sandeep Kumar
- Laboratory of Cancer Immunology, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia.
| |
Collapse
|
3
|
Piao M, Feng G. The deubiquitinating enzyme
USP37
promotes keloid fibroblasts proliferation and collagen production by regulating the
c‐Myc
expression. Int Wound J 2022; 20:1517-1524. [PMID: 36333840 PMCID: PMC10088848 DOI: 10.1111/iwj.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/09/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Previous research testifies that c-Myc may promote keloid fibroblast proliferation and collagen accumulation. Ubiquitin-specific peptidase 37 (USP37)-mediated deubiquitination and stabilisation of c-Myc are vital for lung cancer proliferation, while the potential role of USP37 in keloid fibroblasts is not investigated. Elevated USP37, c-Myc, and Collagen I content were detected in keloid tissue with RT-PCR or ELISA assay. USP37 over-expression plasmids or USP37 short hairpin RNAs (shRNAs) were transfected into keloid fibroblasts with Lipofectamine 3000 to decipher the role of USP37 in keloid fibroblasts. USP37 overexpression could promote the proliferation of keloid fibroblasts with increased c-Myc and Collagen I expression. On the other hand, USP37 shRNAs inhibited the proliferation of keloid fibroblasts with diminished c-Myc and Collagen I expression. It was worth noting that C-Myc overexpression promoted the proliferation of keloid fibroblasts inhibited by USP37 shRNAs with increasing Collagen I expression. All of these results demonstrate that USP37 could regulate c-Myc to promote the proliferation and collagen deposit of keloid fibroblasts, and USP37 could be targeted in future keloid therapy.
Collapse
Affiliation(s)
- Meishan Piao
- Department of dermatology the Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University Wuxi China
| | - Guangdong Feng
- Department of dermatology the Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University Wuxi China
| |
Collapse
|
4
|
Crizotinib Resistance Mediated by Autophagy Is Higher in the Stem-Like Cell Subset in ALK-Positive Anaplastic Large Cell Lymphoma, and This Effect Is MYC-Dependent. Cancers (Basel) 2021; 13:cancers13020181. [PMID: 33430343 PMCID: PMC7825760 DOI: 10.3390/cancers13020181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Previously it was shown that autophagy contributes to crizotinib resistance in ALK-positive anaplastic large cell lymphoma (ALK + ALCL). We asked if autophagy is equally important in two distinct subsets of ALK + ALCL, namely Reporter Unresponsive (RU) and Reporter Responsive (RR), of which RR cells display stem-like properties. Autophagic flux was assessed with a fluorescence tagged LC3 reporter and immunoblots to detect endogenous LC3 alongside chloroquine, an autophagy inhibitor. The stem-like RR cells displayed significantly higher autophagic response upon crizotinib treatment. Their exaggerated autophagic response is cytoprotective against crizotinib, as inhibition of autophagy using chloroquine or shRNA against BECN1 or ATG7 led to a decrease in their viability. In contrast, autophagy inhibition in RU resulted in minimal changes. Since the differential protein expression of MYC is a regulator of the RU/RR dichotomy and is higher in RR cells, we asked if MYC regulates the autophagy-mediated cytoprotective effect. Inhibition of MYC in RR cells using shRNA significantly blunted crizotinib-induced autophagic response and effectively suppressed this cytoprotective effect. In conclusion, stem-like RR cells respond with rapid and intense autophagic flux which manifests with crizotinib resistance. For the first time, we have highlighted the direct role of MYC in regulating autophagy and its associated chemoresistance phenotype in ALK + ALCL stem-like cells.
Collapse
|
5
|
El-Rashid M, Ghimire K, Sanganeria B, Lu B, Rogers NM. CD47 limits autophagy to promote acute kidney injury. FASEB J 2019; 33:12735-12749. [PMID: 31480863 DOI: 10.1096/fj.201900120rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Acute kidney injury (AKI) initiates a complex pathophysiological cascade leading to epithelial cell death. Recent studies identify autophagy, a key intracellular process that degrades cytoplasmic constituents, as protective against AKI. We have previously reported that the protein thrombospondin-1 and its receptor CD47 are induced in AKI; however, the mechanism underlying their regulation of injury is unknown. Here, we investigated whether CD47 signaling affects autophagy to regulate AKI. Wild-type (WT) and CD47-/- mice were challenged with renal ischemia-reperfusion injury. All animals underwent analysis of renal function and biomolecular phenotyping. CD47-/- mice were resistant to AKI, with decreased serum creatinine and ameliorated histologic changes compared with WT animals. These mice also displayed increased abundance of key autophagy genes, including autophagy-related gene (Atg)5, Atg7, beclin-1, and microtubule-associated proteins 1A/1B light chain 3 (LC3) at baseline and post-AKI, which were significantly reduced in WT mice. Changes in protein expression correlated with increased autophagosome and autolysosome formation in renal tubular epithelial cells (RTECs). In mouse kidney transplantation, treatment with a CD47-blocking antibody that improved function was associated with increased autophagy compared with control mice. Primary isolated RTECs from CD47-/- mice demonstrated increased basal expression of several autophagy components that was preserved under hypoxic stress. These data suggest that activated CD47 promotes AKI through inhibition of autophagy and point to CD47 as a target to preserve renal function following injury.-El-Rashid, M., Ghimire, K., Sanganeria, B., Lu, B., Rogers, N. M. CD47 limits autophagy to promote acute kidney injury.
Collapse
Affiliation(s)
- Maryam El-Rashid
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Kedar Ghimire
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Barkha Sanganeria
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Bo Lu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Natasha M Rogers
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia.,Renal Division, Westmead Hospital, Westmead, New South Wales, Australia.,Westmead Clinical Medical School, University of Sydney, Camperdown, New South Wales, Australia.,Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
6
|
Riyahi N, Safaroghli-Azar A, Sheikh-Zeineddini N, Sayyadi M, Bashash D. Synergistic Effects of PI3K and c-Myc Co-targeting in Acute Leukemia: Shedding New Light on Resistance to Selective PI3K-δ Inhibitor CAL-101. Cancer Invest 2019; 37:311-324. [DOI: 10.1080/07357907.2019.1651328] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Niknam Riyahi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ava Safaroghli-Azar
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Sheikh-Zeineddini
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Sayyadi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Liu L, Liao JZ, He XX, Li PY. The role of autophagy in hepatocellular carcinoma: friend or foe. Oncotarget 2017; 8:57707-57722. [PMID: 28915706 PMCID: PMC5593678 DOI: 10.18632/oncotarget.17202] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved lysosome-dependent catabolic process which degrades cell’s components in order to recycle substrates to exert optimally and adapt to tough circumstances. It is a critical cellular homeostatic mechanism with stress resistance, immunity, antiaging, and pro-tumor or anti-tumor effects. Among these, the role of autophagy in cancer is the most eye-catching that is not immutable but dynamic and highly complex. Basal autophagy acts as a tumor suppressor by maintaining genomic stability in normal cells. However, once a tumor is established, unbalanced autophagy will contribute to carcinoma cell survival under tumor microenvironment and in turn promote tumor growth and development. The dynamic role of autophagy can also apply on hepatocellular carcinoma (HCC). HCC is a highly malignant cancer with high morbidity and poor survival rate. Decline or overexpression of autophagic essential genes such as ATG7, ATG5 or Beclin 1 plays a key role in the occurrence and development of HCC but the exact mechanisms are still highly controversial. Signaling pathways or molecules involving in autophagy, for example PI3K/AKT/mTOR pathway, ERK/MAPK pathway, PERK pathway, p53, LncRNA PTENP1 (Long non-coding RNA PTENP1), microRNA-375 and so on, occupy an important position in the complex role of autophagy in HCC. Here, we discuss the dynamic role, the signaling pathways and the potential prognostic and therapy value of autophagy in HCC.
Collapse
Affiliation(s)
- Lian Liu
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Zhi Liao
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-Xing He
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei-Yuan Li
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Belounis A, Nyalendo C, Le Gall R, Imbriglio TV, Mahma M, Teira P, Beaunoyer M, Cournoyer S, Haddad E, Vassal G, Sartelet H. Autophagy is associated with chemoresistance in neuroblastoma. BMC Cancer 2016; 16:891. [PMID: 27846885 PMCID: PMC5109645 DOI: 10.1186/s12885-016-2906-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 10/27/2016] [Indexed: 12/19/2022] Open
Abstract
Background Neuroblastoma (NB) is a frequent pediatric tumor characterized by a poor prognosis where a majority of tumors progress despite intensive multimodality treatments. Autophagy, a self-degradative process in cells, could be induced by chemotherapy and be associated with chemoresistance. The aim of this study was to determine whether: 1) autophagy is present in NB, 2) chemotherapy modified its levels, and 3) its inhibition decreased chemoresistance. Methods Immunohistochemical stainings were performed on samples from 184 NB patients in order to verify the expression of LC3B, a specific marker for autophagy, and Beclin 1, a positive regulator of autophagy. In addition, we performed an in vitro study with six NB cell lines and six drugs (vincristine, doxorubicin, cisplatin temozolomide, LY294002 and syrolimus). Inhibition of autophagy was performed using ATG5 knockdown cells or hydroxychloroquine (HCQ). Cell survival was measured using the MTT cell proliferation assay. Autophagy was detected by monodansylcadaverine, confocal microscopy and Western blot. In vivo study with tumor xenografts in NSG mice was performed. Results Our results have indicated that autophagy was present at low levels in NB and was not a prognostic factor, while Beclin 1 was highly expressed in children with poor NB prognosis. However, autophagy levels increased after chemotherapy in vitro and in vivo. Tumor progression was significantly decreased in mice treated with a combination of HCQ and vincristine. Conclusions Taken together, autophagy is present in NB, induced by chemotherapy and associated with chemoresistance, which is significantly reduced by its inhibition. Therefore, targeting autophagy represents a very attractive approach to develop new therapeutic strategies in NB. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2906-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Assila Belounis
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada.,Department of pathology and cellular biology, Université de Montréal, Montreal, QC, Canada
| | - Carine Nyalendo
- Department of biochemistry, CHU Sainte Justine, Montreal, QC, Canada
| | - Roxane Le Gall
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada
| | - Tina V Imbriglio
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada
| | - Mohamed Mahma
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada
| | - Pierre Teira
- Division of paediatric oncology, CHU Sainte Justine, Montreal, QC, Canada
| | - Mona Beaunoyer
- Department of surgery, CHU Sainte Justine, 3175, Montreal, QC, Canada
| | - Sonia Cournoyer
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada
| | - Elie Haddad
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada
| | - Gilles Vassal
- Department of paediatric oncology, Institut Gustave Roussy, Villejuif, France
| | - Hervé Sartelet
- Research centre of the Sainte Justine university hospital, Montreal, QC, Canada. .,Department of pathology and cellular biology, Université de Montréal, Montreal, QC, Canada. .,Department of pathology and cytogenetic, CHU Sainte Justine, Montreal, QC, Canada.
| |
Collapse
|
9
|
Jacomin AC, Samavedam S, Promponas V, Nezis IP. iLIR database: A web resource for LIR motif-containing proteins in eukaryotes. Autophagy 2016; 12:1945-1953. [PMID: 27484196 PMCID: PMC5079668 DOI: 10.1080/15548627.2016.1207016] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Atg8-family proteins are the best-studied proteins of the core autophagic machinery. They are essential for the elongation and closure of the phagophore into a proper autophagosome. Moreover, Atg8-family proteins are associated with the phagophore from the initiation of the autophagic process to, or just prior to, the fusion between autophagosomes with lysosomes. In addition to their implication in autophagosome biogenesis, they are crucial for selective autophagy through their ability to interact with selective autophagy receptor proteins necessary for the specific targeting of substrates for autophagic degradation. In the past few years it has been revealed that Atg8-interacting proteins include not only receptors but also components of the core autophagic machinery, proteins associated with vesicles and their transport, and specific proteins that are selectively degraded by autophagy. Atg8-interacting proteins contain a short linear LC3-interacting region/LC3 recognition sequence/Atg8-interacting motif (LIR/LRS/AIM) motif which is responsible for their interaction with Atg8-family proteins. These proteins are referred to as LIR-containing proteins (LIRCPs). So far, many experimental efforts have been carried out to identify new LIRCPs, leading to the characterization of some of them in the past 10 years. Given the need for the identification of LIRCPs in various organisms, we developed the iLIR database ( https://ilir.warwick.ac.uk ) as a freely available web resource, listing all the putative canonical LIRCPs identified in silico in the proteomes of 8 model organisms using the iLIR server, combined with a Gene Ontology (GO) term analysis. Additionally, a curated text-mining analysis of the literature permitted us to identify novel putative LICRPs in mammals that have not previously been associated with autophagy.
Collapse
Affiliation(s)
| | - Siva Samavedam
- a School of Life Sciences, University of Warwick , Coventry , UK
| | - Vasilis Promponas
- b Bioinformatics Research Laboratory, Department of Biological Sciences, University of Cyprus , Nicosia , Cyprus
| | - Ioannis P Nezis
- a School of Life Sciences, University of Warwick , Coventry , UK
| |
Collapse
|
10
|
Corrado M, Scorrano L, Campello S. Changing perspective on oncometabolites: from metabolic signature of cancer to tumorigenic and immunosuppressive agents. Oncotarget 2016; 7:46692-46706. [PMID: 27083002 PMCID: PMC5216830 DOI: 10.18632/oncotarget.8727] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/31/2016] [Indexed: 12/12/2022] Open
Abstract
During tumorigenesis, the shift from oxidative phosphorylation to glycolysis in ATP production accounts for the dramatic change in the cellular metabolism and represents one of the major steps leading to tumour formation. The so-called Warburg effect is currently considered something more than a mere modification in the cellular metabolism. The paradox that during cancer cell proliferation the increase in energy need is supplied by glycolysis can be only explained by taking into account the many roles that intermediates of glycolysis or TCA cycle play in cellular physiology, besides energy production. Recent studies have shown that metabolic intermediates induce changes in chromatin structure or drive neo-angiogenesis. In this review, we present some of the latest findings in the study of cancer metabolism with particular attention to how tumour metabolism and its microenvironment can favour tumour growth and aggressiveness, by hijacking and dampening the anti-tumoral immune response.
Collapse
Affiliation(s)
- Mauro Corrado
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy
- IRCCS Fondazione Santa Lucia, Roma, Italy
| | - Luca Scorrano
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, Padova, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Silvia Campello
- IRCCS Fondazione Santa Lucia, Roma, Italy
- Department of Biology, University of Roma Tor Vergata, Roma, Italy
| |
Collapse
|
11
|
Li Z, Hao S, Yin H, Gao J, Yang Z. Autophagy ameliorates cognitive impairment through activation of PVT1 and apoptosis in diabetes mice. Behav Brain Res 2016; 305:265-77. [PMID: 26971628 DOI: 10.1016/j.bbr.2016.03.023] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 02/24/2016] [Accepted: 03/09/2016] [Indexed: 01/19/2023]
Abstract
The underlying mechanisms of cognitive impairment in diabetes remain incompletely characterized. Here we show that the autophagic inhibition by 3-methyladenine (3-MA) aggravates cognitive impairment in streptozotocin-induced diabetic mice, including exacerbation of anxiety-like behaviors and aggravation in spatial learning and memory, especially the spatial reversal memory. Further neuronal function identification confirmed that both long term potentiation (LTP) and depotentiation (DPT) were exacerbated by autophagic inhibition in diabetic mice, which indicating impairment of synaptic plasticity. However, no significant change of pair-pulse facilitation (PPF) was recorded in diabetic mice with autophagic suppression compared with the diabetic mice, which indicated that presynaptic function was not affected by autophagic inhibition in diabetes. Subsequent hippocampal neuronal cell death analysis showed that the apoptotic cell death, but not the regulated necrosis, significantly increased in autophagic suppression of diabetic mice. Finally, molecular mechanism that may lead to cell death was identified. The long non-coding RNA PVT1 (plasmacytoma variant translocation 1) expression was analyzed, and data revealed that PVT1 was decreased significantly by 3-MA in diabetes. These findings show that PVT1-mediated autophagy may protect hippocampal neurons from impairment of synaptic plasticity and apoptosis, and then ameliorates cognitive impairment in diabetes. These intriguing findings will help pave the way for exciting functional studies of autophagy in cognitive impairment and diabetes that may alter the existing paradigms.
Collapse
Affiliation(s)
- Zhigui Li
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Animal Models and Degenerative Neurological Diseases, Nankai University, Tianjin 300071, China
| | - Shuang Hao
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Animal Models and Degenerative Neurological Diseases, Nankai University, Tianjin 300071, China
| | - Hongqiang Yin
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Animal Models and Degenerative Neurological Diseases, Nankai University, Tianjin 300071, China
| | - Jing Gao
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Animal Models and Degenerative Neurological Diseases, Nankai University, Tianjin 300071, China
| | - Zhuo Yang
- College of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Animal Models and Degenerative Neurological Diseases, Nankai University, Tianjin 300071, China.
| |
Collapse
|
12
|
Autophagy and mTORC1 regulate the stochastic phase of somatic cell reprogramming. Nat Cell Biol 2015; 17:715-25. [PMID: 25985393 DOI: 10.1038/ncb3172] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 03/27/2015] [Indexed: 12/13/2022]
Abstract
We describe robust induction of autophagy during the reprogramming of mouse fibroblasts to induced pluripotent stem cells by four reprogramming factors (Sox2, Oct4, Klf4 and c-Myc), henceforth 4F. This process occurs independently of p53 activation, and is mediated by the synergistic downregulation of mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy-related genes. The 4F coordinately repress mTORC1, but bifurcate in their regulation of autophagy-related genes, with Klf4 and c-Myc inducing them but Sox2 and Oct4 inhibiting them. On one hand, inhibition of mTORC1 facilitates reprogramming by promoting cell reshaping (mitochondrial remodelling and cell size reduction). On the other hand, mTORC1 paradoxically impairs reprogramming by triggering autophagy. Autophagy does not participate in cell reshaping in reprogramming but instead degrades p62, whose accumulation in autophagy-deficient cells facilitates reprogramming. Our results thus reveal a complex signalling network involving mTORC1 inhibition and autophagy induction in the early phase of reprogramming, whose delicate balance ultimately determines reprogramming efficiency.
Collapse
|
13
|
Brindle NR, Joyce JA, Rostker F, Lawlor ER, Swigart-Brown L, Evan G, Hanahan D, Shchors K. Deficiency for the cysteine protease cathepsin L impairs Myc-induced tumorigenesis in a mouse model of pancreatic neuroendocrine cancer. PLoS One 2015; 10:e0120348. [PMID: 25927437 PMCID: PMC4415914 DOI: 10.1371/journal.pone.0120348] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/21/2015] [Indexed: 02/06/2023] Open
Abstract
Motivated by the recent implication of cysteine protease cathepsin L as a potential target for anti-cancer drug development, we used a conditional MycERTAM;Bcl-xL model of pancreatic neuroendocrine tumorigenesis (PNET) to assess the role of cathepsin L in Myc-induced tumor progression. By employing a cysteine cathepsin activity probe in vivo and in vitro, we first established that cathepsin activity increases during the initial stages of MycERTAM;Bcl-xL tumor development. Among the cathepsin family members investigated, only cathepsin L was predominately produced by beta-tumor cells in neoplastic pancreata and, consistent with this, cathepsin L mRNA expression was rapidly upregulated following Myc activation in the beta cell compartment. By contrast, cathepsins B, S and C were highly enriched in tumor-infiltrating leukocytes. Genetic deletion of cathepsin L had no discernible effect on the initiation of neoplastic growth or concordant angiogenesis. However, the tumors that developed in the cathepsin L-deficient background were markedly reduced in size relative to their typical wild-type counterparts, indicative of a role for cathepsin L in enabling expansive tumor growth. Thus, genetic blockade of cathepsin L activity is inferred to retard Myc-driven tumor growth, encouraging the potential utility of pharmacological inhibitors of cysteine cathepsins in treating late stage tumors.
Collapse
Affiliation(s)
- Nicola R. Brindle
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Johanna A. Joyce
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Fanya Rostker
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
| | - Elizabeth R. Lawlor
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
| | - Lamorna Swigart-Brown
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
| | - Gerard Evan
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
| | - Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Ksenya Shchors
- Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
- Departments of Pathology and Department of Biochemistry and Biophysics, University of California San Francisco (UCSF), San Francisco, United States of America
| |
Collapse
|
14
|
Therapeutic targeting of autophagy in cancer. Part I: molecular pathways controlling autophagy. Semin Cancer Biol 2014; 31:89-98. [PMID: 24879905 DOI: 10.1016/j.semcancer.2014.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/09/2014] [Accepted: 05/18/2014] [Indexed: 12/31/2022]
Abstract
Autophagy is a process in which cells can generate energy and building materials, by degradation of redundant and/or damaged organelles and proteins. Especially during conditions of stress, autophagy helps to maintain homeostasis. In addition, autophagy has been shown to influence malignant transformation and cancer progression. The precise molecular events in autophagy are complex and the core autophagic machinery described to date consists of nearly thirty proteins. Apart from these factors that execute the process of autophagy, several signalling pathways are involved in converting internal and external stimuli into an autophagic response. In this review we provide an overview of the signalling pathways that influence autophagy, particularly in cancer cells. We will illustrate that interference with multiple of these signalling pathways can have significant effects on cancer cell survival.
Collapse
|
15
|
Hiroi N, Swat M, Funahashi A. Assessing uncertainty in model parameters based on sparse and noisy experimental data. Front Physiol 2014; 5:128. [PMID: 24772089 PMCID: PMC3983526 DOI: 10.3389/fphys.2014.00128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 03/14/2014] [Indexed: 11/13/2022] Open
Abstract
To perform parametric identification of mathematical models of biological events, experimental data are rare to be sufficient to estimate target behaviors produced by complex non-linear systems. We performed parameter fitting to a cell cycle model with experimental data as an in silico experiment. We calibrated model parameters with the generalized least squares method with randomized initial values and checked local and global sensitivity of the model. Sensitivity analyses showed that parameter optimization induced less sensitivity except for those related to the metabolism of the transcription factors c-Myc and E2F, which are required to overcome a restriction point (R-point). We performed bifurcation analyses with the optimized parameters and found the bimodality was lost. This result suggests that accumulation of c-Myc and E2F induced dysfunction of R-point. We performed a second parameter optimization based on the results of sensitivity analyses and incorporating additional derived from recent in vivo data. This optimization returned the bimodal characteristics of the model with a narrower range of hysteresis than the original. This result suggests that the optimized model can more easily go through R-point and come back to the gap phase after once having overcome it. Two parameter space analyses showed metabolism of c-Myc is transformed as it can allow cell bimodal behavior with weak stimuli of growth factors. This result is compatible with the character of the cell line used in our experiments. At the same time, Rb, an inhibitor of E2F, can allow cell bimodal behavior with only a limited range of stimuli when it is activated, but with a wider range of stimuli when it is inactive. These results provide two insights; biologically, the two transcription factors play an essential role in malignant cells to overcome R-point with weaker growth factor stimuli, and theoretically, sparse time-course data can be used to change a model to a biologically expected state.
Collapse
Affiliation(s)
- Noriko Hiroi
- Systems Biology Laboratory, Department of Bioscience and Informatics, Keio University Yokohama, Japan
| | - Maciej Swat
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hermjakob Team: Proteomics Cambridgeshire, UK
| | - Akira Funahashi
- Systems Biology Laboratory, Department of Bioscience and Informatics, Keio University Yokohama, Japan
| |
Collapse
|
16
|
Rapamycin induces apoptosis when autophagy is inhibited in T-47D mammary cells and both processes are regulated by Phlda1. Cell Biochem Biophys 2014; 66:567-87. [PMID: 23300026 DOI: 10.1007/s12013-012-9504-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation pathway and plays a critical role in the homeostatic process of recycling proteins and organelles. Functional relationships have been described between apoptosis and autophagy. Perturbations in the apoptotic machinery have been reported to induce autophagic cell deaths. Inhibition of autophagy in cancer cells has resulted in cell deaths that manifested hallmarks of apoptosis. However, the molecular relationships and the circumstances of which molecular pathways dictate the choice between apoptosis and autophagy are currently unknown. This study aims to identify specific gene expression of rapamycin-induced autophagy and the effects of rapamycin when the autophagy process is inhibited. In this study, we have demonstrated that rapamycin is capable of inducing autophagy in T-47D breast carcinoma cells. However, when the autophagy process was inhibited by 3-MA, the effects of rapamycin became apoptotic. The Phlda1 gene was found to be up-regulated in both autophagy and apoptosis and silencing this gene was found to reduce both activities, strongly suggests that Phlda1 mediates and positively regulates both autophagy and apoptosis pathways.
Collapse
|
17
|
Toh PPC, Luo S, Menzies FM, Raskó T, Wanker EE, Rubinsztein DC. Myc inhibition impairs autophagosome formation. Hum Mol Genet 2013; 22:5237-48. [PMID: 23933736 PMCID: PMC3842180 DOI: 10.1093/hmg/ddt381] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Autophagy, a major clearance route for many long-lived proteins and organelles, has long been implicated in cancer development. Myc is a proto-oncogene often found to be deregulated in many cancers, and thus is an attractive target for design of cancer therapy. Therefore, understanding the relationship between anti-Myc strategies and autophagy will be important for development of effective therapy. Here, we show that Myc depletion inhibits autophagosome formation and impairs clearance of autophagy substrates. Myc suppression has an inhibitory effect on autophagy via reduction of c-Jun N-terminal kinase 1 (JNK1) and B-cell lymphoma 2 (Bcl2) phosphorylation. Additionally, the decrease in JNK1 phosphorylation observed with Myc knockdown is associated with a reduction in ROS production. Our data suggest that targeting Myc in cancer therapy might have the additional benefit of inhibiting autophagy in the case of therapy resistance associated with chemotherapy-induced autophagy.
Collapse
Affiliation(s)
- Pearl P C Toh
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical Research, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
Autophagy is a tightly controlled self-degradation process utilised by cells to sustain cellular homeostasis and to support cell survival in response to metabolic stress and starvation. Thus, autophagy plays a critical role in promoting cell integrity and maintaining proper function of cellular processes. Defects in autophagy, however, can have drastic implications in human health and diseases, including cancer. Described as a double-edged sword in the context of cancer, autophagy can act as both suppressor and facilitator of tumorigenesis. As such, defining the precise role of autophagy in a multistep event like cancer progression can be complex. Recent findings have implicated a role for components of the autophagy pathway in oncogene-mediated cell transformation, tumour growth, and survival. Notably, aggressive cancers driven by Ras oncoproteins rely on autophagy to sustain a reprogrammed mitochondrial metabolic signature and evade cell death. In this review, we summarize our current understanding of the role of oncogene-induced autophagy in cancer progression and discuss how modulators of autophagic responses can bring about therapeutic benefit and eradication of a subset of cancers that are addicted to this ancient recycling machinery.
Collapse
|
19
|
Bif-1 haploinsufficiency promotes chromosomal instability and accelerates Myc-driven lymphomagenesis via suppression of mitophagy. Blood 2013; 121:1622-32. [PMID: 23287860 DOI: 10.1182/blood-2012-10-459826] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Malignant transformation by oncogenes requires additional genetic/epigenetic changes to overcome enhanced susceptibility to apoptosis. In the present study, we report that Bif-1 (Sh3glb1), a gene encoding a membrane curvature–driving endophilin protein, is a haploinsufficient tumor suppressor that plays a key role in the prevention of chromosomal instability and suppresses the acquisition of apoptosis resistance during Myc-driven lymphomagenesis. Although a large portion of Bif-1–deficient mice harboring an Eμ-Myc transgene displayed embryonic lethality, allelic loss of Bif-1 dramatically accelerated the onset of Myc-induced lymphoma. At the premalignant stage, hemizygous deletion of Bif-1 resulted in an increase in mitochondrial mass, accumulation of DNA damage, and up-regulation of the antiapoptotic protein Mcl-1. Consistently, allelic loss of Bif-1 suppressed the activation of caspase-3 in Myc-induced lymphoma cells. Moreover, we found that Bif-1 is indispensable for the autophagy-dependent clearance of damaged mitochondria (mitophagy), because loss of Bif-1 resulted in the accumulation of endoplasmic reticulum–associated immature autophagosomes and suppressed the maturation of autophagosomes. The results of the present study indicate that Bif-1 haploinsufficiency attenuates mitophagy and results in the promotion of chromosomal instability, which enables tumor cells to efficiently bypass the oncogenic/metabolic pressures for apoptosis. .
Collapse
|
20
|
Wang C, Tai Y, Lisanti MP, Liao DJ. c-Myc induction of programmed cell death may contribute to carcinogenesis: a perspective inspired by several concepts of chemical carcinogenesis. Cancer Biol Ther 2011; 11:615-26. [PMID: 21278493 DOI: 10.4161/cbt.11.7.14688] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The c-Myc protein, encoded by c-myc gene, in its wild-type form can induce tumors with a high frequency and can induce massive programmed cell death (PCD) in most transgenic mouse models, with greater efficiency than other oncogenes. Evidence also indicates that c-Myc can cause proliferative inhibition, i.e. mitoinhibition. The c-Myc-induced PCD and mitoinhibition, which may be attributable to its inhibition of cyclin D1 and induction of p53, may impose a pressure of compensatory proliferation, i.e. regeneration, onto the initiated cells (cancer progenitor cells) that occur sporadically and are resistant to the mitoinhibition. The initiated cells can thus proliferate robustly and progress to a malignancy. This hypothetical thinking, i.e. the concurrent PCD and mitoinhibition induced by c-Myc can promote carcinogenesis, predicts that an optimal balance is achieved between cell death and ensuing regeneration during oncogenic transformation by c-Myc, which can better promote carcinogenesis. In this perspective, we summarize accumulating evidence and challenge the current model that oncoprotein induces carcinogenesis by promoting cellular proliferation and/or inhibiting PCD. Inspired by c-myc oncogene, we surmise that many tumor-suppressive or growth-inhibitory genes may also be able to promote carcinogenesis in a similar way, i.e. by inducing PCD and/or mitoinhibition of normal cells to create a need for compensatory proliferation that drives a robust replication of initiating cells.
Collapse
Affiliation(s)
- Chenguang Wang
- Department of Stem Cell and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
21
|
Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci 2010; 36:30-8. [PMID: 20728362 DOI: 10.1016/j.tibs.2010.07.007] [Citation(s) in RCA: 988] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 07/19/2010] [Accepted: 07/20/2010] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are small and highly reactive molecules that can oxidize proteins, lipids and DNA. When tightly controlled, ROS serve as signaling molecules by modulating the activity of the oxidized targets. Accumulating data point to an essential role for ROS in the activation of autophagy. Be the outcome of autophagy survival or death and the initiation conditions starvation, pathogens or death receptors, ROS are invariably involved. The nature of this involvement, however, remains unclear. Moreover, although connections between ROS and autophagy are observed in diverse pathological conditions, the mode of activation of autophagy and its potential protective role remain incompletely understood. Notably, recent advances in the field of redox regulation of autophagy focus on the role of mitochondria as a source of ROS and on mitophagy as a means for clearance of ROS.
Collapse
Affiliation(s)
- Ruth Scherz-Shouval
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | |
Collapse
|
22
|
Tan YK, Kusuma CM, St John LJ, Vu HA, Alibek K, Wu A. Induction of autophagy by anthrax lethal toxin. Biochem Biophys Res Commun 2008; 379:293-7. [PMID: 19103170 DOI: 10.1016/j.bbrc.2008.12.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 12/10/2008] [Indexed: 12/19/2022]
Abstract
Autophagy is an evolutionary conserved intracellular process whereby cells break down long-lived proteins and organelles. Accumulating evidences suggest increasing physiological significance of autophagy in pathogenesis of infectious diseases. Anthrax lethal toxin (LT) exerts its influence on numerous cells and herein, we report a novel effect of LT-induced autophagy on mammalian cells. Several autophagy biochemical markers including LC3-II conversion, increased punctuate distribution of GFP-LC3 and development of acidic vesicular organelles (AVO) were detected in cells treated with LT. Analysis of individual LT component revealed a moderate increase in LC3-II conversion for protective antigen-treated cells, whereas the LC3-II level in lethal factor-treated cells remained unchanged. In addition, our preliminary findings suggest a protective role of autophagy in LT intoxication as autophagy inhibition resulted in accelerated cell death. This study presents a hitherto undescribed effect of LT-induced autophagy on cells and provides the groundwork for future studies on the implication of autophagy in anthrax pathogenesis.
Collapse
Affiliation(s)
- Yian Kim Tan
- Molecular and Microbiology Department, College of Sciences, George Mason University, Manassas, VA 22010, USA
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
A host of dietary factors can influence various cellular processes and thereby potentially influence overall cancer risk and tumor behavior. In many cases, these factors suppress cancer by stimulating programmed cell death. However, death not only can follow the well-characterized type I apoptotic pathway but also can proceed by nonapoptotic modes such as type II (macroautophagy-related) and type III (necrosis) or combinations thereof. In contrast to apoptosis, the induction of macroautophagy may contribute to either the survival or death of cells in response to a stressor. This review highlights current knowledge and gaps in our understanding of the interactions among bioactive food constituents, autophagy, and cancer. Whereas a variety of food components including vitamin D, selenium, curcumin, resveratrol, and genistein have been shown to stimulate autophagy vacuolization, it is often difficult to determine if this is a protumorigenic or antitumorigenic response. Additional studies are needed to examine dose and duration of exposures and tissue specificity in response to bioactive food components in transgenic and knockout models to resolve the physiologic implications of early changes in the autophagy process.
Collapse
Affiliation(s)
- Keith Singletary
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA.
| | | |
Collapse
|
24
|
Polager S, Ofir M, Ginsberg D. E2F1 regulates autophagy and the transcription of autophagy genes. Oncogene 2008; 27:4860-4. [PMID: 18408756 DOI: 10.1038/onc.2008.117] [Citation(s) in RCA: 201] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The retinoblastoma pathway is often inactivated in human tumors resulting in deregulated E2F activity that can induce both proliferation and cell death. Although the role of E2F in apoptosis is well characterized, little is known regarding its putative participation in other cell death pathways. We show here that activation of E2F1 upregulates the expression of four autophagy genes-microtubule-associated protein-1 light chain-3 (LC3), autophagy-related gene-1 (ATG1), ATG5 and damage-regulated autophagy modulator (DRAM). E2F1-mediated induction of LC3, ATG1 and DRAM is direct and indeed, endogenous E2F1 can be found bound to regions encompassing the promoters of these genes. Regulation of ATG5 by E2F1 is indirect. Importantly, we demonstrate that E2F1 activation enhances autophagy and conversely, reducing endogenous E2F1 expression inhibits DNA damage-induced autophagy. These studies identify E2F1 as a transcriptional regulator of autophagy, and for the first time establish a role for E2F1 in DNA damage-induced autophagy.
Collapse
Affiliation(s)
- S Polager
- The Mina and Everard Goodman Faculty of Life Science, Bar Ilan University, Ramat Gan, Israel
| | | | | |
Collapse
|
25
|
Cui Q, Tashiro SI, Onodera S, Ikejima T. Augmentation of oridonin-induced apoptosis observed with reduced autophagy. J Pharmacol Sci 2007; 101:230-9. [PMID: 16861822 DOI: 10.1254/jphs.fpj06003x] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Our previous studies showed that oridonin could induce apoptosis in HeLa cells; and in this study, we further investigated autophagy induced by oridonin in HeLa cells and the relationship between apoptosis and autophagy. HeLa cells were exposed with oridonin after 3-methyladenine (3-MA) pre-culture, and we evaluated the growth inhibitory ratio, morphologic changes, DNA fragmentation, proteins expression as well as autophagic and apoptotic levels. Oridonin inhibited the proliferation of HeLa cells in vitro and induced autophagy. Oligonucleosomal fragementation of DNA as well as increased activities of Bax proteins were induced by oridonin, but the expression of p-Bcl-2 protein was reduced. In the condition of oridonin-treatment, when the inhibitor of phosphoinositide 3-kinase (PI3K), wortmannin, was applied, the autophagic level was significantly decreased, while the apoptotic level was increased, indicating that PI3K is a key regulator of both autophagy and apoptosis. Akt, down-stream factor of PI3K, was activated in autophagic process but suppressed in apoptosis in this study. In addition, when autophagy was blocked by 3-MA, the expression of SIRT-1 was decreased, indicating SIRT-1 contributed to autophagy. Taken together, oridonin simultaneously induced HeLa cell both apoptosis and autophagy in HeLa cells, and inhibition of autophagy contributes to upregulation of apoptosis.
Collapse
Affiliation(s)
- Qiao Cui
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, China
| | | | | | | |
Collapse
|
26
|
Afanasyeva EA, Komarova EY, Larsson LG, Bahram F, Margulis BA, Guzhova IV. Drug-induced Myc-mediated apoptosis of cancer cells is inhibited by stress protein Hsp70. Int J Cancer 2007; 121:2615-21. [PMID: 17694514 DOI: 10.1002/ijc.22974] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The Myc oncoprotein serves a dual function by stimulating cells both towards growth and apoptosis. The latter functions are often abrogated during tumor development. The Hsp70 stress protein is a potent anti-apoptotic molecule, but its potential role in protecting cells from Myc-mediated apoptosis has not been investigated. Our results show that activated Myc potentiated apoptosis induced by the cancer drugs etoposide (ETO) and camptothecin (CAMP) in v-Myc-expressing human U-937 monoblastic cells and in Rat1 cells containing a conditionally active Myc/estrogen receptor (MycER) fusion protein. However, both heat shock and ectopic Hsp70 expression protected the cells from Myc-mediated apoptosis after drug treatment in both systems. The increased susceptibility to the anti-tumor drugs by activated Myc was enhanced by siRNA-mediated knockdown of Hsp70 expression in U-937 cells. Addressing the mechanisms by which Myc and Hsp70 promotes and inhibits drug-induced apoptosis, respectively, we found that v-Myc stimulated cytochrome c release and activation of effector caspase-9, -3 and -7, but not of initiator caspase-8. Inhibition of caspase-9 specifically reduced v-Myc-stimulated apoptosis, whereas inhibition of caspase-8 and -3/7 reduced apoptosis both in v-myc-expressing and parental ETO-treated U-937 cells. Interestingly, Myc-stimulated activation of effector caspases was inhibited, but cytochrome c release was not affected by Hsp70 expression, suggesting that Hsp70 interferes with the proapoptotic function of Myc downstream of mitochondria, at the level of caspase-9 and downstream caspases. In conclusion, Hsp70 seems to have key function in inhibition of apoptosis mediated by Myc and may therefore play an important role in Myc-driven oncogenesis.
Collapse
Affiliation(s)
- Elena A Afanasyeva
- Laboratory of Cell Protection Mechanism, Institute of Cytology RAS, St. Petersburg 194064, Russia
| | | | | | | | | | | |
Collapse
|
27
|
Lum JJ, DeBerardinis RJ, Thompson CB. Autophagy in metazoans: cell survival in the land of plenty. Nat Rev Mol Cell Biol 2005; 6:439-48. [PMID: 15928708 DOI: 10.1038/nrm1660] [Citation(s) in RCA: 605] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cells require a constant supply of macromolecular precursors and oxidizable substrates to maintain viability. Unicellular eukaryotes lack the ability to regulate nutrient concentrations in their extracellular environment. So when environmental nutrients are depleted, these organisms catabolize existing cytoplasmic components to support ATP production to maintain survival, a process known as autophagy. By contrast, the environment of metazoans normally contains abundant extracellular nutrients, but a cell's ability to take up these nutrients is controlled by growth factor signal transduction. Despite evolving the ability to maintain a constant supply of extracellular nutrients, metazoans have retained a complete set of autophagy genes. The physiological relevance of autophagy in such species is just beginning to be explored.
Collapse
Affiliation(s)
- Julian J Lum
- Abramson Family Cancer Research Institute, Department of Cancer Biology, University of Pennsylvania School of Medicine, Philadelphia, Philadelphia 19104, USA
| | | | | |
Collapse
|
28
|
Meijer AJ, Codogno P. Regulation and role of autophagy in mammalian cells. Int J Biochem Cell Biol 2005; 36:2445-62. [PMID: 15325584 DOI: 10.1016/j.biocel.2004.02.002] [Citation(s) in RCA: 443] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2003] [Revised: 01/16/2004] [Accepted: 02/05/2004] [Indexed: 01/25/2023]
Abstract
The recent period has witnessed progress in the understanding of the lysosomal autophagic pathway. The discovery of a family of genes conserved from yeast to humans, and involved in the formation of autophagosomes, has unraveled new protein-conjugation systems and has shed light on the importance of autophagy in physiology and pathophysiology. The elucidation of the molecular control of autophagy will also lead to a better understanding of the role of autophagy during cell death. As a great number of extracellular stimuli (starvation, hormonal or therapeutic treatment) as well as intracellular stimuli (accumulation of misfolded proteins, invasion of microorganisms) is able to modulate the autophagic response, it is not surprising that several signaling pathways are involved in the control of autophagy. The mammalian Target of Rapamycin (mTOR) signaling pathway plays a major role in transmitting autophagic stimuli because of its ability to sense nutrient, metabolic and hormonal signals. In addition, autophagy, which is characterized by a flux of membrane from the formation of the autophagosome to the fusion with the lysosome, is regulated by GTPases, similarly to the vesicular transport along the exocytic/endocytic pathway. The aim of the present review is to give an overview of autophagy and to discuss its regulation by activators and effectors of mTOR and GTPases.
Collapse
Affiliation(s)
- Alfred J Meijer
- Department of Biochemistry, Academic Medical Center, University of Amsterdam, Meibergreef 15, 1105 AZ Amsterdam, The Netherlands
| | | |
Collapse
|
29
|
Affiliation(s)
- Hitoshi Okada
- Institute for Breast Cancer Research/Ontario Cancer Institute, 620 University Avenue, Toronto, Ontario, Canada M5G 2C1
| | | |
Collapse
|
30
|
Abstract
Autophagy is characterized by sequestration of bulk cytoplasm and organelles in double or multimembrane autophagic vesicles, and their delivery to and subsequent degradation by the cell's own lysosomal system. Autophagy has multiple physiological functions in multicellular organisms, including protein degradation and organelle turnover. Genes and proteins that constitute the basic machinery of the autophagic process were first identified in the yeast system and some of their mammalian orthologues have been characterized as well. Increasing lines of evidence indicate that these molecular mechanisms may be recruited by an alternative, caspase-independent form of programmed cell death, named autophagic type II cell death. In some settings, autophagy and apoptosis seem to be interconnected positively or negatively, introducing the concept of 'molecular switches' between them. Additionally, mitochondria may be central organelles integrating the two types of cell death. Malignant transformation is frequently associated with suppression of autophagy. The recent implication of tumor suppressors like Beclin 1, DAP-kinase and PTEN in autophagic pathways indicates a causative role for autophagy deficiencies in cancer formation. Autophagic cell death induction by some anticancer agents underlines the potential utility of its induction as a new cancer treatment modality.
Collapse
Affiliation(s)
- Devrim Gozuacik
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|