1
|
Gao Y, Tang Y. Emerging roles of prohibitins in cancer: an update. Cancer Gene Ther 2025; 32:357-370. [PMID: 40057573 DOI: 10.1038/s41417-025-00883-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 04/09/2025]
Abstract
The prohibitin (PHB) family, including PHB1 and its homolog PHB2, is ubiquitously located in different cellular compartments and plays roles in fundamental cellular processes such as proliferation, differentiation, and apoptosis. Accumulating evidence has indicated that this family contributes to the development of numerous diseases in particular cancers. Aberrant expressions of PHBs can been observed in diverse types of human cancer. Depending on their cell compartment-specific attributes and interacting proteins, PHBs are tightly linked to almost all aspects of cancer biology and have distinct bidirectional functions of tumor-suppression or tumor-promotion. However, the roles of PHBs in cancer have yet to be fully characterized and understood. This review provides an updated overview of the pleiotropic effects of PHBs and emphasizes their characteristic roles in each cancer respectively, with the great expectation to identify potential targets for therapeutic approaches and promising molecular biomarkers for cancer diagnosis and prognostic monitor.
Collapse
Affiliation(s)
- Yunliang Gao
- Department of Urology, the Second Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Mental Disorders, Changsha, China
- Hunan Clinical Research Center of Minimally Invasive Urology, Changsha, China
| | - Yuanyuan Tang
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
2
|
Jo H, Kim M, Jeoung J, Kim W, Park YH, Jung HS, Lee W, Jeoung D. Rocaglamide Suppresses Allergic Reactions by Regulating IL-4 Receptor Signaling. Molecules 2025; 30:840. [PMID: 40005151 PMCID: PMC11858170 DOI: 10.3390/molecules30040840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Rocaglamide (Roc-A), a natural phytochemical isolated from Aglaia species, is known to exert anticancer effects. Allergic inflammation can enhance the tumorigenic potential of cancer cells. We hypothesized that Roc-A could regulate allergic inflammation. Roc-A prevented an antigen from increasing the hallmarks of allergic reactions in vitro. Roc-A suppressed passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). RNA sequencing analysis showed that Roc-A prevented the antigen from increasing the expression of IL-4 in RBL2H3 cells. Roc-A also prevented the antigen from increasing the expression of interleukin-4 receptor (IL-4R). Roc-A was found to form a hydrogen-bonding network with residues N92 and L64 of IL-4R in a molecular docking simulation. Roc-A prevented the antigen from inducing the binding of IL-4R to JAK1. Chromatin immunoprecipitation (ChIP) assays showed that C-Jun could bind to promoter sequences of IL-4 and IL-4R. Mouse recombinant IL-4 protein increased β-hexosaminidase activity, IL-4R expression, and the hallmarks of allergic inflammation in the antigen-independent manner. Mouse recombinant IL-4 protein increased the expressions of CD163 and arghinase-1 and markers of M2 macrophages, but decreased the expression of iNOS, a marker of M1 macrophages in lung macrophages. Roc-A regulated the effects of a culture medium of antigen-stimulated RBL2H3 cells on the expressions of iNOS and arginase-1 in RAW264.7 macrophages. The blocking of IL-4 or downregulation of IL-4R exerted negative effects on the hallmarks of allergic reactions in vitro. The blocking of IL-4 or downregulation of IL-4R also exerted negative effects on PCA, and the downregulation of IL-4R exerted negative effects on PSA. An miR-34a mimic exerted negative effects on allergic reactions in vitro. The downregulation of IL-4R prevented the antigen from decreasing the expression of miR-34a in RBL2H3 cells. We identified chemicals that could bind to IL-4R via molecular docking analysis. The IL-4R docking chemical 1536801 prevented the antigen from increasing β-hexosaminidase activity and the hallmarks of allergic reactions. The IL-4R docking chemical 1536801 also exerted a negative effect on PCA. TargetScan analysis predicted miR-34a as a negative regulator of IL-4R. We found that the anti-allergic effect of Roc-A and its mechanisms were associated with miR-34a. Taken together, our results show that understanding IL-4R-mediated allergic reactions can provide clues for the development of anti-allergy therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, Republic of Korea; (H.J.); (M.K.); (J.J.); (W.K.); (H.S.J.); (W.L.)
| |
Collapse
|
3
|
Valcikova B, Vadovicova N, Smolkova K, Zacpalova M, Krejci P, Lee S, Rauch J, Kolch W, von Kriegsheim A, Dorotikova A, Andrysik Z, Vichova R, Vacek O, Soucek K, Uldrijan S. eIF4F controls ERK MAPK signaling in melanomas with BRAF and NRAS mutations. Proc Natl Acad Sci U S A 2024; 121:e2321305121. [PMID: 39436655 PMCID: PMC11536119 DOI: 10.1073/pnas.2321305121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/12/2024] [Indexed: 10/23/2024] Open
Abstract
The eIF4F translation initiation complex plays a critical role in melanoma resistance to clinical BRAF and MEK inhibitors. In this study, we uncover a function of eIF4F in the negative regulation of the rat sarcoma (RAS)/rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) signaling pathway. We demonstrate that eIF4F is essential for controlling ERK signaling intensity in treatment-naïve melanoma cells harboring BRAF or NRAS mutations. Specifically, the dual-specificity phosphatase DUSP6/MKP3, which acts as a negative feedback regulator of ERK activity, requires continuous production in an eIF4F-dependent manner to limit excessive ERK signaling driven by oncogenic RAF/RAS mutations. Treatment with small-molecule eIF4F inhibitors disrupts the negative feedback control of MAPK signaling, leading to ERK hyperactivation and EGR1 overexpression in melanoma cells in vitro and in vivo. Furthermore, our quantitative analyses reveal a high spare signaling capacity in the ERK pathway, suggesting that eIF4F-dependent feedback keeps the majority of ERK molecules inactive under normal conditions. Overall, our findings highlight the crucial role of eIF4F in regulating ERK signaling flux and suggest that pharmacological eIF4F inhibitors can disrupt the negative feedback control of MAPK activity in melanomas with BRAF and NRAS activating mutations.
Collapse
Affiliation(s)
- Barbora Valcikova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
| | - Natalia Vadovicova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
| | - Karolina Smolkova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
| | - Magdalena Zacpalova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
- Laboratory of Cell Signaling, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Brno60200, Czech Republic
| | - Shannon Lee
- Systems Biology Ireland, School of Medicine, University College Dublin, DublinD04 V1W8, Ireland
| | - Jens Rauch
- Systems Biology Ireland, School of Medicine, University College Dublin, DublinD04 V1W8, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, DublinD04 V1W8, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, DublinD04 V1W8, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, DublinD04 V1W8, Ireland
| | - Alexander von Kriegsheim
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, EdinburghEH4 2XR, United Kingdom
| | - Anna Dorotikova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
| | - Zdenek Andrysik
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045
| | - Rachel Vichova
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno61200, Czech Republic
| | - Ondrej Vacek
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno61200, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno62500, Czech Republic
| | - Karel Soucek
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno61200, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno62500, Czech Republic
| | - Stjepan Uldrijan
- Department of Biology, Faculty of Medicine, Masaryk University, Brno62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno60200, Czech Republic
| |
Collapse
|
4
|
Krieger MR, Abrahamian M, He KL, Atamdede S, Hakimjavadi H, Momcilovic M, Ostrow D, Maggo SD, Tsang YP, Gai X, Chanfreau GF, Shackelford DB, Teitell MA, Koehler CM. Trafficking of mitochondrial double-stranded RNA from mitochondria to the cytosol. Life Sci Alliance 2024; 7:e202302396. [PMID: 38955468 PMCID: PMC11220484 DOI: 10.26508/lsa.202302396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
In addition to mitochondrial DNA, mitochondrial double-stranded RNA (mtdsRNA) is exported from mitochondria. However, specific channels for RNA transport have not been demonstrated. Here, we begin to characterize channel candidates for mtdsRNA export from the mitochondrial matrix to the cytosol. Down-regulation of SUV3 resulted in the accumulation of mtdsRNAs in the matrix, whereas down-regulation of PNPase resulted in the export of mtdsRNAs to the cytosol. Targeting experiments show that PNPase functions in both the intermembrane space and matrix. Strand-specific sequencing of the double-stranded RNA confirms the mitochondrial origin. Inhibiting or down-regulating outer membrane proteins VDAC1/2 and BAK/BAX or inner membrane proteins PHB1/2 strongly attenuated the export of mtdsRNAs to the cytosol. The cytosolic mtdsRNAs subsequently localized to large granules containing the stress protein TIA-1 and activated the type 1 interferon stress response pathway. Abundant mtdsRNAs were detected in a subset of non-small-cell lung cancer cell lines that were glycolytic, indicating relevance in cancer biology. Thus, we propose that mtdsRNA is a new damage-associated molecular pattern that is exported from mitochondria in a regulated manner.
Collapse
Affiliation(s)
- Matthew R Krieger
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | | | - Kevin L He
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Sean Atamdede
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | | | - Milica Momcilovic
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Dejerianne Ostrow
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Simran Ds Maggo
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yik Pui Tsang
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Xiaowu Gai
- Department of Pathology, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guillaume F Chanfreau
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - David B Shackelford
- Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
| | - Michael A Teitell
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center, UCLA, Los Angeles, CA, USA
- NanoSystems Institute, UCLA, Los Angeles, CA, USA
| | - Carla M Koehler
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
5
|
Najem A, Krayem M, Sabbah S, Pesetti M, Journe F, Awada A, Désaubry L, Ghanem GE. Targeting Prohibitins to Inhibit Melanoma Growth and Overcome Resistance to Targeted Therapies. Cells 2023; 12:1855. [PMID: 37508519 PMCID: PMC10378173 DOI: 10.3390/cells12141855] [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: 05/10/2023] [Revised: 06/19/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Despite important advances in the treatment of metastatic melanoma with the development of MAPK-targeted agents and immune checkpoint inhibitors, the majority of patients either do not respond to therapies or develop acquired resistance. Furthermore, there is no effective targeted therapy currently available for BRAF wild-type melanomas (approximately 50% of cutaneous melanoma). Thus, there is a compelling need for new efficient targeted therapies. Prohibitins (PHBs) are overexpressed in several types of cancers and implicated in the regulation of signaling networks that promote cell invasion and resistance to cell apoptosis. Herein, we show that PHBs are highly expressed in melanoma and are associated with not only poor survival but also with resistance to BRAFi/MEKi. We designed and identified novel specific PHB inhibitors that can inhibit melanoma cell growth in 3D spheroid models and a large panel of representative cell lines with different molecular subtypes, including those with intrinsic and acquired resistance to MAPKi, by significantly moderating both MAPK (CRAF-ERK axis) and PI3K/AKT pathways, and inducing apoptosis through the mitochondrial pathway and up-regulation of p53. In addition, autophagy inhibition enhances the antitumor efficacy of these PHB ligands. More important, these ligands can act in synergy with MAPKi to more efficiently inhibit cell growth and overcome drug resistance in both BRAF wild-type and mutant melanoma. In conclusion, targeting PHBs represents a very promising therapeutic strategy in melanoma, regardless of mutational status.
Collapse
Affiliation(s)
- Ahmad Najem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Mohammad Krayem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Serena Sabbah
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Matilde Pesetti
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Fabrice Journe
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| | - Ahmad Awada
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Laurent Désaubry
- Center of Research in Biomedicine of Strasbourg, Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, 67000 Strasbourg, France
| | - Ghanem E Ghanem
- Laboratory of Clinical and Experimental Oncology (LOCE), Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium
| |
Collapse
|
6
|
Liu J, Zhang R, Su T, Zhou Q, Gao L, He Z, Wang X, Zhao J, Xing Y, Sun F, Cai W, Wang X, Han J, Qin R, Désaubry L, Han B, Chen W. Targeting PHB1 to inhibit castration-resistant prostate cancer progression in vitro and in vivo. J Exp Clin Cancer Res 2023; 42:128. [PMID: 37210546 DOI: 10.1186/s13046-023-02695-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/01/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Castration-resistant prostate cancer (CRPC) is currently the main challenge for prostate cancer (PCa) treatment, and there is an urgent need to find novel therapeutic targets and drugs. Prohibitin (PHB1) is a multifunctional chaperone/scaffold protein that is upregulated in various cancers and plays a pro-cancer role. FL3 is a synthetic flavagline drug that inhibits cancer cell proliferation by targeting PHB1. However, the biological functions of PHB1 in CRPC and the effect of FL3 on CRPC cells remain to be explored. METHODS Several public datasets were used to analyze the association between the expression level of PHB1 and PCa progression as well as outcome in PCa patients. The expression of PHB1 in human PCa specimens and PCa cell lines was examined by immunohistochemistry (IHC), qRT-PCR, and Western blot. The biological roles of PHB1 in castration resistance and underlying mechanisms were investigated by gain/loss-of-function analyses. Next, in vitro and in vivo experiments were conducted to investigate the anti-cancer effects of FL3 on CRPC cells as well as the underlying mechanisms. RESULTS PHB1 expression was significantly upregulated in CRPC and was associated with poor prognosis. PHB1 promoted castration resistance of PCa cells under androgen deprivation condition. PHB1 is an androgen receptor (AR) suppressive gene, and androgen deprivation promoted the PHB1 expression and its nucleus-cytoplasmic translocation. FL3, alone or combined with the second-generation anti-androgen Enzalutamide (ENZ), suppressed CRPC cells especially ENZ-sensitive CRPC cells both in vitro and in vivo. Mechanically, we demonstrated that FL3 promoted trafficking of PHB1 from plasma membrane and mitochondria to nucleus, which in turn inhibited AR signaling as well as MAPK signaling, yet promoted apoptosis in CRPC cells. CONCLUSION Our data indicated that PHB1 is aberrantly upregulated in CRPC and is involved in castration resistance, as well as providing a novel rational approach for treating ENZ-sensitive CRPC.
Collapse
Affiliation(s)
- Junmei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ranran Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tong Su
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianqian Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zongyue He
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jian Zhao
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanxin Xing
- Research Center of Basic Medicine, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Feifei Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinpei Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingying Han
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ruixi Qin
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China
| | - Laurent Désaubry
- INSERM, UMR 1260, Regenerative Nanomedicine, University of Strasbourg, FMTS (Fédération de Médecine Translationnelle de L'Université de Strasbourg), Strasbourg, France
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Department of Pathology, Qilu Hospital of Shandong University, Jinan, China.
| | - Weiwen Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
7
|
Sánchez-Vera I, Núñez-Vázquez S, Saura-Esteller J, Cosialls AM, Heib J, Nadal Rodríguez P, Ghashghaei O, Lavilla R, Pons G, Gil J, Iglesias-Serret D. The Prohibitin-Binding Compound Fluorizoline Activates the Integrated Stress Response through the eIF2α Kinase HRI. Int J Mol Sci 2023; 24:ijms24098064. [PMID: 37175767 PMCID: PMC10179266 DOI: 10.3390/ijms24098064] [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: 03/24/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Fluorizoline is a synthetic molecule that induces apoptosis, by selectively targeting prohibitins (PHBs), through induction of the BH3-only protein NOXA. This induction is transcriptionally regulated by the integrated stress response (ISR)-related transcription factors ATF3 and ATF4. Here, we evaluate the role of the four eIF2α kinases, to decipher which is responsible for the mechanism of ISR activation triggered by fluorizoline in HeLa and HAP1 cells. First, we demonstrated the involvement of the eIF2α kinases using ISR inhibitor (ISRIB) and by simultaneous downregulation of all four eIF2α kinases, as both approaches were able to increase cell resistance to fluorizoline-induced apoptosis. Furthermore, we confirmed that fluorizoline treatment results in endoplasmic reticulum (ER) stress, as evidenced by PERK activation. Despite PERK activation, this kinase was not directly involved in the ISR activation by fluorizoline. In this regard, we found that the eIF2α kinases are capable of compensating for each other's loss of function. Importantly, we demonstrated that the mitochondrial-stress-related eIF2α kinase HRI mediates ISR activation after fluorizoline treatment.
Collapse
Affiliation(s)
- Ismael Sánchez-Vera
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Ana M Cosialls
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Judith Heib
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Pau Nadal Rodríguez
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Ouldouz Ghashghaei
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Rodolfo Lavilla
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), University of Barcelona, 08028 Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), 08907 L'Hospitalet de Llobregat, Spain
| | - Daniel Iglesias-Serret
- Departament d'Infermeria Fonamental i Medicoquirúrgica, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08907 L'Hospitalet de Llobregat, Spain
- Facultat de Medicina, Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), 08500 Vic, Spain
| |
Collapse
|
8
|
Qi A, Lamont L, Liu E, Murray SD, Meng X, Yang S. Essential Protein PHB2 and Its Regulatory Mechanisms in Cancer. Cells 2023; 12:cells12081211. [PMID: 37190120 DOI: 10.3390/cells12081211] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Prohibitins (PHBs) are a highly conserved class of proteins and have an essential role in transcription, epigenetic regulation, nuclear signaling, mitochondrial structural integrity, cell division, and cellular membrane metabolism. Prohibitins form a heterodimeric complex, consisting of two proteins, prohibitin 1 (PHB1) and prohibitin 2 (PHB2). They have been discovered to have crucial roles in regulating cancer and other metabolic diseases, functioning both together and independently. As there have been many previously published reviews on PHB1, this review focuses on the lesser studied prohibitin, PHB2. The role of PHB2 in cancer is controversial. In most human cancers, overexpressed PHB2 enhances tumor progression, while in some cancers, it suppresses tumor progression. In this review, we focus on (1) the history, family, and structure of prohibitins, (2) the essential location-dependent functions of PHB2, (3) dysfunction in cancer, and (4) the promising modulators to target PHB2. At the end, we discuss future directions and the clinical significance of this common essential gene in cancer.
Collapse
Affiliation(s)
- Amanda Qi
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Lillie Lamont
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Evelyn Liu
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Sarina D Murray
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Xiangbing Meng
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Shujie Yang
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
9
|
Zhang X, Zhao J, Li Q, Qin D, Li W, Wang X, Bi M, Li Q, Li T. Lamprey prohibitin 2 inhibits non-small cell lung carcinoma cell proliferation by down-regulating the expression and phosphorylation levels of cell cycle-associated proteins. FISH & SHELLFISH IMMUNOLOGY 2023; 134:108560. [PMID: 36681363 DOI: 10.1016/j.fsi.2023.108560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/31/2022] [Accepted: 01/18/2023] [Indexed: 06/17/2023]
Abstract
Prohibitin 2 (PHB2) is an evolutionarily conserved and functionally diverse protein that plays an important role in multiple cellular functions, including cell proliferation, cell migration, and apoptosis, and is also known to participate in the process of tumorigenesis and development. In this study, the lamprey PHB2 (Lm-PHB2) gene was over-expressed in KRAS (kirsten rat sarcoma viral oncogene homolog)-mutated non-small cell lung carcinoma (NSCLC) cells to investigate its effect on cell proliferation. The effects of Lm-PHB2 protein on the proliferation of NSCLC cells were determined by treating cells with the purified recombinant Lm-PHB2 protein (rLm-PHB2) followed by cell counting kit (CCK) assays and flow cytometry. Analysis showed that rLm-PHB2 blocked cells in the G2 phase and inhibited the cell proliferation of A549, Calu-1, and NCI-H226 to various degrees. The effect on Calu-1 cells was the most obvious and was concentration- and time-dependent. Similarly, cells transfected with the pEGFP-N1-Lm-PHB2 plasmid also resulted in the suppression of proliferation in A549 cells and Calu-1 cells. Quantitative real-time polymerase chain reaction (qRT-PCR) showed that Lm-PHB2 inhibited cell proliferation by repressing the transcription of PLK1 (polo-like kinase 1), Wee1 (wee1 kinase), CCNB1 (cyclin B1), and CDC25C (cell division control protein 25C). According to western blot analysis, Lm-PHB2 not only down-regulated the expression of PLK1, Wee1, CCNB1, and CDC25C but also reduced the phosphorylation levels of CCNB1 and CDC25C, thus blocking Calu-1 cells in G2/M phase. Our findings demonstrate a function of lamprey PHB2 that may inhibit the proliferation of some NSCLC cells by down-regulating the expression and phosphorylation of cell cycle-associated proteins.
Collapse
Affiliation(s)
- Xue Zhang
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Jianzhu Zhao
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Qing Li
- School of Science and Engineering, University of Dundee, Dundee, DD1 5EN, UK
| | - Di Qin
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Wenwei Li
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Xinyu Wang
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Mengfei Bi
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China
| | - Qingwei Li
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116023, China
| | - Tiesong Li
- College of Life Sciences, Lamprey Research Center, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, 116081, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116023, China.
| |
Collapse
|
10
|
Non-classical ferroptosis inhibition by a small molecule targeting PHB2. Nat Commun 2022; 13:7473. [PMID: 36463308 PMCID: PMC9719519 DOI: 10.1038/s41467-022-35294-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
Ferroptosis is a new type of programmed cell death characterized by iron-dependent lipid peroxidation. Ferroptosis inhibition is thought as a promising therapeutic strategy for a variety of diseases. Currently, a majority of known ferroptosis inhibitors belong to either antioxidants or iron-chelators. Here we report a new ferroptosis inhibitor, termed YL-939, which is neither an antioxidant nor an iron-chelator. Chemical proteomics revealed the biological target of YL-939 to be prohibitin 2 (PHB2). Mechanistically, YL-939 binding to PHB2 promotes the expression of the iron storage protein ferritin, hence reduces the iron content, thereby decreasing the susceptibility to ferroptosis. We further showed that YL-939 could substantially ameliorate liver damage in a ferroptosis-related acute liver injury model by targeting the PHB2/ferritin/iron axis. Overall, we identified a non-classical ferroptosis inhibitor and revealed a new regulation mechanism of ferroptosis. These findings may present an attractive intervention strategy for ferroptosis-related diseases.
Collapse
|
11
|
Development of fluorizoline analogues as prohibitin ligands that modulate C-RAF signaling, p21 expression and melanogenesis. Eur J Med Chem 2022; 242:114635. [DOI: 10.1016/j.ejmech.2022.114635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
|
12
|
Mu H, Zeng Y, Zhuang Y, Gao W, Zhou Y, Rajalingam K, Zhao W. Patterning of Oncogenic Ras Clustering in Live Cells Using Vertically Aligned Nanostructure Arrays. NANO LETTERS 2022; 22:1007-1016. [PMID: 35044178 DOI: 10.1021/acs.nanolett.1c03886] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
As a dominant oncogenic protein, Ras is well-known to segregate into clusters on the plasma membrane for activating downstream signaling. However, current technologies for direct measurements of Ras clustering are limited to sophisticated high-resolution techniques like electron microscopy and fluorescence lifetime imaging. To further promote fundamental investigations and the related drug development, we hereby introduce a nanobar-based platform which effectively guides Ras clusters into quantifiable patterns in live cells that is resolvable under conventional microscopy. Major Ras isoforms, K-Ras, H-Ras, and N-Ras, were differentiated, as well as their highly prevalent oncogenic mutants G12V and G13D. Moreover, the isoform specificity and the sensitivity of a Ras inhibitor were successfully characterized on nanobars. We envision that this nanobar-based platform will serve as an effective tool to read Ras clustering on the plasma membrane, enabling a novel avenue both to decipher Ras regulations and to facilitate anti-Ras drug development.
Collapse
Affiliation(s)
- Huanwen Mu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
- Ageing Research Institute for Society and Education, Nanyang Technological University, Singapore 637335, Singapore
| | - Yongpeng Zeng
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Yinyin Zhuang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| | - Weibo Gao
- Division of Physics and Applied Physics, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore 637371, Singapore
- The Photonics Institute and Centre for Disruptive Photonic Technologies, Nanyang Technological University, Singapore 637371, Singapore
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences Center, Houston, Texas 77030, United States
- Graduate Program in Biochemistry and Cell Biology, MD Anderson Cancer Center and UTHealth Graduate School of Biomedical Sciences, Houston, Texas 77030, United States
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
- University Cancer Center Mainz, University Medical Center Mainz, Johannes Gutenberg University, D 55131 Mainz, Germany
| | - Wenting Zhao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore
| |
Collapse
|
13
|
Takagi H, Moyama C, Taniguchi K, Ando K, Matsuda R, Ando S, Ii H, Kageyama S, Kawauchi A, Chouha N, Désaubry L, Nakata S. Fluorizoline Blocks the Interaction between Prohibitin-2 and γ-Glutamylcyclotransferase and Induces p21Waf1/Cip1 Expression in MCF7 Breast Cancer Cells. Mol Pharmacol 2022; 101:78-86. [PMID: 34862308 DOI: 10.1124/molpharm.121.000334] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022] Open
Abstract
Prohibitin-2 (PHB2) is a scaffold protein that has pleiotropic functions, which include interacting with γ-glutamylcyclotransferase (GGCT) in the cytoplasm and repressing the transcriptional activities of the p21Waf1/Cip (p21) gene in the nucleus. The cytotoxic drug fluorizoline binds to PHB1/2 and exerts antiproliferative actions on cancer cells. However, the precise mechanism underlying the antiproliferative effects of fluorizoline is not fully elucidated. In the present study, we first show that fluorizoline induces p21 expression in several human cancer cell lines, including MCF7 breast cancer cells. Treatment of MCF7 cells with fluorizoline suppressed proliferation and prevented cells from entering into the DNA synthesis phase. Knockdown of p21 rescued the suppressed proliferation, indicating that fluorizoline inhibited MCF7 cell growth via the induction of p21. Overexpression of PHB2 in MCF7 cells prevented the induction of p21 expression by fluorizoline and restored the antiproliferative effects and blockade of cell cycle progression. Moreover, treatment of MCF7 cells with fluorizoline inhibited the interaction between endogenous PHB2 and GGCT proteins and reduced the level of nuclear localization of PHB2 proteins. These results indicate that targeting PHB2 with fluorizoline induces the expression of p21 and consequently blocks proliferation of cancer cells. SIGNIFICANCE STATEMENT: This study shows that fluorizoline may be a promising novel anticancer drug candidate that induces p21 expression and blocks cell-cycle progression in human cancer cell lines. In addition, we show that fluorizoline inhibits the interaction between PHB2 and GGCT and reduces the nuclear localization of PHB2 proteins.
Collapse
Affiliation(s)
- Hiroko Takagi
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Chiami Moyama
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Keiko Taniguchi
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Kota Ando
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Ryohei Matsuda
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Shota Ando
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Hiromi Ii
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Susumu Kageyama
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Akihiro Kawauchi
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Nora Chouha
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Laurent Désaubry
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| | - Susumu Nakata
- Department of Clinical Oncology, Kyoto Pharmaceutical University, Kyoto, Japan (H.T., C.M., K.T., K.A., R.M., S.A., H.I., S.N.); Department of Urology, Shiga University of Medical Science, Otsu, Japan (S.K., A.K.); University of Batna 2, Faculty of Biology, Batna, Algeria (N.C.); Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, INSERM-University of Strasbourg, Strasbourg, France (L.D.); and Sino-French Joint Laboratory of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China (L.D.)
| |
Collapse
|
14
|
Yan X, Yao C, Fang C, Han M, Gong C, Hu D, Shen W, Wang L, Li S, Zhu S. Rocaglamide promotes the infiltration and antitumor immunity of NK cells by activating cGAS-STING signaling in non-small cell lung cancer. Int J Biol Sci 2022; 18:585-598. [PMID: 35002511 PMCID: PMC8741839 DOI: 10.7150/ijbs.65019] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Natural killer (NK) cell-based immunotherapy is clinically limited due to insufficient tumor infiltration in solid tumors. We have previously found that the natural product rocaglamide (RocA) can enhance NK cell-mediated killing of non-small cell lung cancer (NSCLC) cells by inhibiting autophagy, and autophagic inhibition has been shown to increase NK cell tumor infiltration in melanoma. Therefore, we hypothesized that RocA could increase NK cell infiltration in NSCLC by autophagy inhibition. Methods: Flow cytometry, RNA-sequencing, real-time PCR, Western blotting analysis, and xenograft tumor model were utilized to assess the infiltration of NK cells and the underlying mechanism. Results: RocA significantly increased the infiltration of NK cells and the expressions of CCL5 and CXCL10 in NSCLC cells, which could not be reversed by the inhibitions of autophagy/ULK1, JNK and NF-κB. However, such up-regulation could be suppressed by the inhibitions of TKB1 and STING. Furthermore, RocA dramatically activated the cGAS (cyclic GMP-AMP synthase)-STING (stimulator of interferon genes) signaling pathway, and the inhibition/depletion of STING ablated the up-regulation of CCL5 and CXCL10, NK cell infiltration, and tumor regression induced by RocA. Besides, RocA damaged mitochondrial DNA (mtDNA) and promoted the cytoplasmic release of mtDNA. The mPTP inhibitor cyclosporin A could reverse RocA-induced cytoplasmic release of mtDNA. Conclusions: RocA could promote NK cell infiltration by activating cGAS-STING signaling via targeting mtDNA, but not by inhibiting autophagy. Taken together, our current findings suggested that RocA was a potent cGAS-STING agonist and had a promising potential in cancer immunotherapy, especially in NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Xuewei Yan
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Chao Yao
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Cheng Fang
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Min Han
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Chenyuan Gong
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Dan Hu
- School of Acupuncture, Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Weiming Shen
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Lixin Wang
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Suyun Li
- Department of Pathology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| | - Shiguo Zhu
- Center for Traditional Chinese Medicine and Immunology Research; School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China.,Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, P. R. China
| |
Collapse
|
15
|
Zhao Y, Wang Y, Chen W, Bai S, Peng W, Zheng M, Yang Y, Cheng B, Luan Z. Targeted intervention of eIF4A1 inhibits EMT and metastasis of pancreatic cancer cells via c-MYC/miR-9 signaling. Cancer Cell Int 2021; 21:670. [PMID: 34906136 PMCID: PMC8672469 DOI: 10.1186/s12935-021-02390-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background Owing to the lack of effective treatment options, early metastasis remains the major cause of pancreatic ductal adenocarcinoma (PDAC) recurrence and mortality. However, the molecular mechanism of early metastasis is largely unknown. We characterized the function of eukaryotic translation initiation factors (eIFs) in epithelial-mesenchymal-transition (EMT) and metastasis in pancreatic cancer cells to investigate whether eIFs and downstream c-MYC affect EMT and metastasis by joint interference. Methods We used The Cancer Genome Atlas (TCGA) and Genome Tissue Expression (GTEx) databases to analyze eIF4A1 expression in PDAC tissues and further validated the findings with a microarray containing 53 PDAC samples. Expression regulation and pharmacological inhibition of eIF4A1 and c-MYC were performed to determine their role in migration, invasion, and metastasis in pancreatic cancer cells in vitro and in vivo. Results Elevated eIF4A1 expression was positively correlated with lymph node infiltration, tumor size, and indicated a poor prognosis. eIF4A1 decreased E-cadherin expression through the c-MYC/miR-9 axis. Loss of eIF4A1 and c-MYC decreased the EMT and metastasis capabilities of pancreatic cancer cells, whereas upregulation of eIF4A1 attenuated the inhibition of EMT and metastasis induced by c-MYC downregulation. Treatment with the eIF4A1 inhibitor rocaglamide (RocA) or the c-MYC inhibitor Mycro3 either alone or in combination significantly decreased the expression level of EMT markers in pancreatic cancer cells in vitro. However, the efficiency and safety of RocA alone were not inferior to those of the combination treatment in vivo. Conclusion Overexpression of eIF4A1 downregulated E-cadherin expression through the c-MYC/miR-9 axis, which promoted EMT and metastasis of pancreatic cancer cells. Despite the potential feedback loop between eIF4A1 and c-MYC, RocA monotherapy is a promising treatment inhibiting eIF4A1-induced PDAC metastasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02390-0.
Collapse
Affiliation(s)
- Yuchong Zhao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Yun Wang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China.,Departement of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Jianshe East Road No. 1, Zhengzhou, China
| | - Wei Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Shuya Bai
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Wang Peng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Mengli Zheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Yilei Yang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China
| | - Bin Cheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China.
| | - Zhou Luan
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue No. 1095, Wuhan, 430030, China.
| |
Collapse
|
16
|
Belser M, Walker DW. Role of Prohibitins in Aging and Therapeutic Potential Against Age-Related Diseases. Front Genet 2021; 12:714228. [PMID: 34868199 PMCID: PMC8636131 DOI: 10.3389/fgene.2021.714228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022] Open
Abstract
A decline in mitochondrial function has long been associated with age-related health decline. Several lines of evidence suggest that interventions that stimulate mitochondrial autophagy (mitophagy) can slow aging and prolong healthy lifespan. Prohibitins (PHB1 and PHB2) assemble at the mitochondrial inner membrane and are critical for mitochondrial homeostasis. In addition, prohibitins (PHBs) have diverse roles in cell and organismal biology. Here, we will discuss the role of PHBs in mitophagy, oxidative phosphorylation, cellular senescence, and apoptosis. We will also discuss the role of PHBs in modulating lifespan. In addition, we will review the links between PHBs and diseases of aging. Finally, we will discuss the emerging concept that PHBs may represent an attractive therapeutic target to counteract aging and age-onset disease.
Collapse
Affiliation(s)
- Misa Belser
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
17
|
Prohibitin, STAT3 and SH2D4A physically and functionally interact in tumor cell mitochondria. Cell Death Dis 2020; 11:1023. [PMID: 33257655 PMCID: PMC7705682 DOI: 10.1038/s41419-020-03220-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022]
Abstract
Chromosome 8p is frequently deleted in various cancer entities and has been shown to correlate with poor patient survival. SH2D4A is located on chromosome 8p and prevents the nuclear translocation of the pro-tumorigenic transcription factor STAT3. Here, we investigated the interaction of SH2D4A and STAT3 to shed light on the non-canonical functions of STAT3 in cooperation with the tumor suppressor SH2D4A. Using an immunoprecipitation-mass spectrometry (IP-MS) approach, we identified the mitochondrial scaffold proteins prohibitin 1 (PHB1) and prohibitin 2 (PHB2) among other proteins to potentially bind to SH2D4A. Co-immunoprecipitation and proximity ligation assays confirmed direct interactions of STAT3, PHB1, and SH2D4A in situ and in vitro. In addition, cell fractionation and immunofluorescence staining revealed co-localization of these proteins with mitochondria. These interactions were selectively interrupted by the small molecule and PHB ligand FL3. Furthermore, FL3 led to a reduction of STAT3 protein levels, STAT3 transcriptional activity, and HIF1α protein stabilization upon dimethyloxalylglycine (DMOG) treatment. Besides, mitochondrial fusion and fission markers, L-OPA1, Mfn1, and FIS1, were dysregulated upon FL3 treatment. This dysregulated morphology was accompanied by significant reduction of mitochondrial respiration, thus, FL3 significantly diminished mitochondrial respirational capacity. In contrast, SH2D4A knockout increased mitochondrial respiration, whereas FL3 reversed the effect of SH2D4A knockout. The here described results indicate that the interaction of SH2D4A and PHB1 is involved in the mitochondrial function and integrity. The demonstrated interaction with STAT3, accompanied by its reduction of transcriptional activity, further suggests that SH2D4A is linking STAT3 to its mitochondrial functions, and inhibition of PHB-interaction may have therapeutic effects in tumor cells with STAT3 activation.
Collapse
|
18
|
Wang D, Tabti R, Elderwish S, Djehal A, Chouha N, Pinot F, Yu P, Nebigil CG, Désaubry L. SFPH proteins as therapeutic targets for a myriad of diseases. Bioorg Med Chem Lett 2020; 30:127600. [PMID: 33035678 PMCID: PMC7536521 DOI: 10.1016/j.bmcl.2020.127600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/27/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022]
Abstract
The stomatin/prohibitin/flotillin/HflK/HflC (SPFH) domain is present in an evolutionarily conserved family of proteins that regulate a myriad of signaling pathways in archaea, bacteria and eukaryotes. The most studied SPFH proteins, prohibitins, have already been targeted by different families of small molecules to induce anticancer, cardioprotective, anti-inflammatory, antiviral, and antiosteoporotic activities. Ligands of other SPFH proteins have also been identified and shown to act as anesthetics, anti-allodynia, anticancer, and anti-inflammatory agents. These findings indicate that modulators of human or bacterial SPFH proteins can be developed to treat a wide variety of human disorders.
Collapse
Affiliation(s)
- Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Redouane Tabti
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Sabria Elderwish
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Amel Djehal
- Superior National School Biotechnology Taoufik Khaznadar, Constantine, Algeria
| | - Nora Chouha
- University of Batna 2, Faculty of Biology, Batna, Algeria
| | - Franck Pinot
- University of Strasbourg, CNRS, IBMP UPR 2357, Strasbourg, France
| | - Peng Yu
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Canan G Nebigil
- INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China; INSERM-University of Strasbourg, Regenerative Nanomedicine Laboratory (UMR1260), Faculty of Medicine, FMTS, Strasbourg, France.
| |
Collapse
|
19
|
Huang X, Liu J, Ma Q. Prohibitin participates in the HIRA complex to promote cell metastasis in breast cancer cell lines. FEBS Open Bio 2020; 10:2182-2190. [PMID: 32865342 PMCID: PMC7530387 DOI: 10.1002/2211-5463.12966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 06/20/2020] [Accepted: 07/20/2020] [Indexed: 12/04/2022] Open
Abstract
Prohibitin (PHB) is a highly conserved, ubiquitously expressed, multifunctional protein with a well‐characterized function as a chaperone‐stabilizing mitochondrial proteins. Recently it was reported that nuclear PHB participates in HIRA chaperone complexes and regulates downstream gene expression via cell cycle independent deposition of H3.3 into DNA. However, the role of PHB in cancer progression remains controversial with conflicting reports in the literature, perhaps due to its cell type‐dependent subcellular localization. Here, we report that the increased expression of nuclear PHB is positively correlated with metastasis of breast cancer cell lines. We showed PHB participates in the HIRA complex by interacting with HIRA through the linker region of the PHB domain and stabilizes all components of the HIRA complex in breast cancer. Overexpression of nuclear PHB resulted in a higher enrichment of histone H3.3 deposited by the HIRA complex at the promoters of mesenchymal markers. This coincided with an increased gene expression level of these markers, and induced EMT in breast cancer. Overall, these molecular and structural mechanisms suggest that nuclear PHB could hold promise as a potential target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqing Huang
- Department of Oncology and Hematology, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), China
| | - Jinji Liu
- Department of Oncology and Hematology, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), China
| | - Qinghui Ma
- Department of Oncology and Hematology, The Second People's Hospital of Foshan (Affiliated Foshan Hospital of Southern Medical University), China
| |
Collapse
|
20
|
Wang D, Tabti R, Elderwish S, Abou-Hamdan H, Djehal A, Yu P, Yurugi H, Rajalingam K, Nebigil CG, Désaubry L. Prohibitin ligands: a growing armamentarium to tackle cancers, osteoporosis, inflammatory, cardiac and neurological diseases. Cell Mol Life Sci 2020; 77:3525-3546. [PMID: 32062751 PMCID: PMC11104971 DOI: 10.1007/s00018-020-03475-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 02/08/2023]
Abstract
Over the last three decades, the scaffold proteins prohibitins-1 and -2 (PHB1/2) have emerged as key signaling proteins regulating a myriad of signaling pathways in health and diseases. Small molecules targeting PHBs display promising effects against cancers, osteoporosis, inflammatory, cardiac and neurodegenerative diseases. This review provides an updated overview of the various classes of PHB ligands, with an emphasis on their mechanism of action and therapeutic potential. We also describe how these ligands have been used to explore PHB signaling in different physiological and pathological settings.
Collapse
Affiliation(s)
- Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Redouane Tabti
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Sabria Elderwish
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Hussein Abou-Hamdan
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Amel Djehal
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
- Superior National School Biotechnology Taoufik Khaznadar, Ville universitaire Ali Mendjeli, BP E66 25100, Constantine, Algeria
| | - Peng Yu
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Hajime Yurugi
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | | | - Canan G Nebigil
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
- Laboratory of Cardio-Oncology and Medicinal Chemistry (FRE 2033), CNRS, Institut Le Bel, 4 rue Blaise Pascal, CS 90032, 67081, Strasbourg, France.
| |
Collapse
|
21
|
Guo WW, Chen LH, Yin W, Mo LX. Aberrant Expression of Prohibitin Is Related to Prognosis of Nasal Extranodal Natural Killer/T Cell Lymphoma, Nasal Type. Oncol Res Treat 2020; 43:491-497. [PMID: 32756049 DOI: 10.1159/000509094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/02/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Nasal extranodal natural killer (NK)/T cell lymphoma, nasal type (ENKTCL) is a high-grade Epstein-Barr virus (EBV)-associated malignancy with poor outcomes. There are few biomarkers for the accurate diagnosis and prognostic prediction of the disease. The aim of this study was to investigate the clinicopathological significance of prohibitin (PHB) expression in nasal ENKTCL. METHODS The expression level of PHB was detected via immunohistochemical staining in 49 nasal ENKTCL tissues and age- and sex-matched controls of 30 nasal mucosa-reactive lymphoid hyperplasia (NRLH) tissues. The correlations between the PHB expression and clinicopathological features of patients with nasal ENKTCL were evaluated. RESULTS The results indicated a significantly decreased expression of PHB in nasal ENKTCL tissues compared with in NRLH tissues. Low-level PHB expression was significantly associated with younger age and fever (p = 0.008 and 0.018, respectively). The Kaplan-Meier analysis showed that the cytoplasm expression level of PHB in nasal ENKTCL was inversely related to overall survival (p = 0.046). CONCLUSIONS PHB may be a potential diagnostic marker and prognostic predictor of nasal ENKTCL.
Collapse
Affiliation(s)
- Wen Wen Guo
- Department of Pathology, People's Hospital of Guangxi Province, Nanning, China
| | - Ling Hui Chen
- Department of Pathology, People's Hospital of Guangxi Province, Nanning, China
| | - Wu Yin
- Department of Pathology, People's Hospital of Guangxi Province, Nanning, China
| | - Lan Xiang Mo
- Department of Pathology, People's Hospital of Guangxi Province, Nanning, China,
| |
Collapse
|
22
|
Zhang L, Tian W, Zhou B. Polymorphisms in Neuronal Growth Regulator 1 and Otoancorin Alternate the Susceptibility to Lung Cancer in Chinese Nonsmoking Females. DNA Cell Biol 2020; 39:1657-1663. [PMID: 32552051 DOI: 10.1089/dna.2020.5654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell adhesion molecules (CAMs) play crucial roles in the genesis and progress of tumor. We investigated the effects of single nucleotide polymorphisms (SNPs) of CAMs, neuronal growth regulator 1 (NEGR1), and Otoancorin (OTOA) on lung cancer susceptibility in Chinese nonsmoking females. Logistic regression and Cox regression analyses were conducted to investigate the effects of SNPs and environmental factors. For rs3102911, genotype TT carriers decreased the risk of lung cancer with an odds ratio (OR) of 0.635. AA genotypes of rs741718 increased the risk of lung cancer with an OR of 3.527. In stratified analysis, genotype AA carriers of rs741718 had a high susceptibility to lung adenocarcinoma compared with GG and AG genotypes. Analyses of association between SNPs and clinical characteristics revealed that rs3102911 as a protective factor and rs741718 as a risk factor influenced the lung cancer occurrence and progression in nonsmoking females.
Collapse
Affiliation(s)
- Ludan Zhang
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Clinical Medicine, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Wen Tian
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Baosen Zhou
- Department of Clinical Epidemiology, First Affiliated Hospital, China Medical University, Shenyang, China.,Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| |
Collapse
|
23
|
Jin X, Xie J, Zabolocki M, Wang X, Jiang T, Wang D, Désaubry L, Bardy C, Proud CG. The prohibitin-binding compound fluorizoline affects multiple components of the translational machinery and inhibits protein synthesis. J Biol Chem 2020; 295:9855-9867. [PMID: 32430400 DOI: 10.1074/jbc.ra120.012979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/12/2020] [Indexed: 01/12/2023] Open
Abstract
Fluorizoline (FLZ) binds to prohibitin-1 and -2 (PHB1/2), which are pleiotropic scaffold proteins known to affect signaling pathways involved in several intracellular processes. However, it is not yet clear how FLZ exerts its effect. Here, we show that exposure of three different human cancer cell lines to FLZ increases the phosphorylation of key translation factors, particularly of initiation factor 2 (eIF2) and elongation factor 2 (eEF2), modifications that inhibit their activities. FLZ also impaired signaling through mTOR complex 1, which also regulates the translational machinery, e.g. through the eIF4E-binding protein 4E-BP1. In line with these findings, FLZ potently inhibited protein synthesis. We noted that the first phase of this inhibition involves very rapid eEF2 phosphorylation, which is catalyzed by a dedicated Ca2+-dependent protein kinase, eEF2 kinase (eEF2K). We also demonstrate that FLZ induces a swift and marked rise in intracellular Ca2+ levels, likely explaining the effects on eEF2. Disruption of normal Ca2+ homeostasis can also induce endoplasmic reticulum stress, and our results suggest that induction of this stress response contributes to the increased phosphorylation of eIF2, likely because of activation of the eIF2-modifying kinase PKR-like endoplasmic reticulum kinase (PERK). We show that FLZ induces cancer cell death and that this effect involves contributions from the phosphorylation of both eEF2 and eIF2. Our findings provide important new insights into the biological effects of FLZ and thus the roles of PHBs, specifically in regulating Ca2+ levels, cellular protein synthesis, and cell survival.
Collapse
Affiliation(s)
- Xin Jin
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Jianling Xie
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Michael Zabolocki
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Human Neurophysiology and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Xuemin Wang
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, Australia
| | - Tao Jiang
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology and School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Dong Wang
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Laurent Désaubry
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.,Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Strasbourg, France
| | - Cedric Bardy
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia.,Laboratory for Human Neurophysiology and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, Australia .,School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
24
|
Elderwish S, Audebrand A, Nebigil CG, Désaubry L. Discovery of 3,3'-pyrrolidinyl-spirooxindoles as cardioprotectant prohibitin ligands. Eur J Med Chem 2019; 186:111859. [PMID: 31735574 DOI: 10.1016/j.ejmech.2019.111859] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 01/08/2023]
Abstract
The scaffold proteins prohibitins-1 and 2 (PHB1/2) play many important roles in coordinating many cell signaling pathways and represent emerging targets in cardiology and oncology. We previously reported that a family of natural products derivatives, flavaglines, binds to PHB1/2 to exert cardioprotectant and anti-cancer effects. However, flavaglines also target the initiation factor of translation eIF4A, which doesn't contribute to cardioprotection and may even induce some adverse effects. Herein, we report the development of a convenient and robust synthesis of the new PHB2 ligand 2'-phenylpyrrolidinyl-spirooxindole, and its analogues. We discovered that these compounds displays cardioprotective effect against doxorubicin mediated cardiotoxicity and uncovered the structural requirement for this activity. We identified in particular some analogues that are more cardioprotectant than flavaglines. Pull-down experiments demonstrated that these compounds bind not only to PHB2 but also PHB1. These novel PHB ligands may provide the basis for the development of new drugs candidates to protect the heart against the adverse effects of anticancer treatments.
Collapse
Affiliation(s)
- Sabria Elderwish
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France
| | - Anaïs Audebrand
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Canan G Nebigil
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Laurent Désaubry
- Laboratory of Medicinal Chemistry and Cardio-oncology, CNRS, 4 rue Blaise Pascal, 67081, Strasbourg, France.
| |
Collapse
|
25
|
Bentayeb H, Aitamer M, Petit B, Dubanet L, Elderwish S, Désaubry L, de Gramont A, Raymond E, Olivrie A, Abraham J, Jauberteau MO, Troutaud D. Prohibitin (PHB) expression is associated with aggressiveness in DLBCL and flavagline-mediated inhibition of cytoplasmic PHB functions induces anti-tumor effects. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:450. [PMID: 31684984 PMCID: PMC6830009 DOI: 10.1186/s13046-019-1440-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Diffuse large B-cell lymphomas (DLBCLs) are aggressive lymphomas accounting for approximately a third of non-Hodgkin lymphomas. Prohibitin 1 (PHB1) and prohibitin 2 (PHB2) are scaffold proteins that promote mitochondria homeostasis and consequently cell survival, but biological functions of cytoplasmic PHBs remain largely unknown in DLBCL. METHODS PHB expression was analyzed in 82 DLBCL biopsies and five DLBCL cell lines by immunohistochemistry (IHC) and Western blotting. Pharmacological inhibition of PHB using the synthetic flavagline FL3 was realized in vitro to gain insight PHB cellular functions. Effects of FL3 on DLBCL cell line viability, apoptosis, C-Raf-ERK-MNK-eIF4E signaling pathway and eIF4F complex formation and activity were evaluated by XTT assay, annexin V-FITC/PI dual staining and Western blotting respectively. Subcutaneous DLBCL xenograft model in SCID mice was also performed to determine in vivo FL3 effect. RESULTS As in DLBCL cell lines, PHB1 and PHB2 were expressed in germinal center B-cell-like (GCB) and activated B-cell-like (ABC) subtypes. In patient samples, high PHB levels were associated with higher serum LDH (PHB1 and PHB2), IPIaa (PHB2), and Ki-67 (PHB2) expression. Higher PHB1 expression tends to be associated with shorter event-free survival (EFS) in patients, especially in male patients. FL3 induced apoptosis of DLBCL cell lines that was associated with inhibition of the ERK-MNK-eIF4E signaling pathway, including aggressive double/triple-hit DLBCL cell lines. This resulted in altered eIF4F complex formation and activity leading to a reduction of Bcl-2 and c-Myc expression levels. Moreover, FL3 strongly downregulated DLBCL cellular levels of Akt protein and AKT mRNA. FL3 antitumor activity was also confirmed in vivo in a murine xenograft model. CONCLUSION Our data indicate that PHB overexpression is associated with markers of tumor aggressiveness in DLBCL, and that targeting PHBs may be a therapeutic option, notably in aggressive subtypes.
Collapse
Affiliation(s)
| | | | - Barbara Petit
- Laboratoire d'Anatomie-Pathologique, CHU de Limoges, Limoges, France
| | | | | | - Laurent Désaubry
- UMR 7203, CNRS - Université Paris Sorbonne, Paris, France.,Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | | | - Eric Raymond
- AFR Oncology, 1 place Paul Verlaine, Boulogne-Billancourt, France.,Groupe Hospitalier Saint-Joseph, Paris, France
| | - Agnès Olivrie
- Service d'Hématologie Clinique, CHU de Limoges, Limoges, France
| | - Julie Abraham
- Service d'Hématologie Clinique, CHU de Limoges, Limoges, France
| | - Marie-Odile Jauberteau
- EA3842, Université de Limoges, Limoges, France.,Service d'Immunologie, CHU Limoges, Limoges, France
| | | |
Collapse
|
26
|
MacArthur IC, Bei Y, Garcia HD, Ortiz MV, Toedling J, Klironomos F, Rolff J, Eggert A, Schulte JH, Kentsis A, Henssen AG. Prohibitin promotes de-differentiation and is a potential therapeutic target in neuroblastoma. JCI Insight 2019; 5:127130. [PMID: 30998507 DOI: 10.1172/jci.insight.127130] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gain of the long arm of chromosome 17 (17q) is a cytogenetic hallmark of high-risk neuroblastoma, yet its contribution to neuroblastoma pathogenesis remains incompletely understood. Combining whole-genome and RNA sequencing of neuroblastomas, we identified the prohibitin (PHB) gene as highly expressed in tumors with 17q gain. High PHB expression correlated with poor prognosis and was associated with loss of gene expression programs promoting neuronal development and differentiation. PHB depletion induced differentiation and apoptosis and slowed cell cycle progression of neuroblastoma cells, at least in part through impaired ERK1/2 activation. Conversely, ectopic expression of PHB was sufficient to increase proliferation of neuroblastoma cells and was associated with suppression of markers associated with neuronal differentiation and favorable neuroblastoma outcome. Thus, PHB is a 17q oncogene in neuroblastoma that promotes tumor cell proliferation, and de-differentiation.
Collapse
Affiliation(s)
- Ian C MacArthur
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Medical Scientist Training Program, Albert Einstein College of Medicine, New York, New York, USA
| | - Yi Bei
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Heathcliff Dorado Garcia
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael V Ortiz
- Department of Pediatrics and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Joern Toedling
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Filippos Klironomos
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Rolff
- Experimental Pharmacology and Oncology, Berlin, Germany
| | - Angelika Eggert
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, Heidelberg, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, Heidelberg, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Alex Kentsis
- Department of Pediatrics and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Departments of Pharmacology, Pediatrics, and Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York, USA
| | - Anton G Henssen
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Consortium, Heidelberg, Germany.,Berlin Institute of Health, Berlin, Germany.,Experimental and Clinical Research Center of the Max Delbrück Center and Charité Berlin, Berlin, Germany
| |
Collapse
|
27
|
Yao C, Ni Z, Gong C, Zhu X, Wang L, Xu Z, Zhou C, Li S, Zhou W, Zou C, Zhu S. Rocaglamide enhances NK cell-mediated killing of non-small cell lung cancer cells by inhibiting autophagy. Autophagy 2018; 14:1831-1844. [PMID: 29969944 PMCID: PMC6135631 DOI: 10.1080/15548627.2018.1489946] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 05/24/2018] [Accepted: 06/11/2018] [Indexed: 01/02/2023] Open
Abstract
Targeting macroautophagy/autophagy is a novel strategy in cancer immunotherapy. In the present study, we showed that the natural product rocaglamide (RocA) enhanced natural killer (NK) cell-mediated lysis of non-small cell lung cancer (NSCLC) cells in vitro and tumor regression in vivo. Moreover, this effect was not related to the NK cell recognition of target cells or expressions of death receptors. Instead, RocA inhibited autophagy and restored the level of NK cell-derived GZMB (granzyme B) in NSCLC cells, therefore increasing their susceptibility to NK cell-mediated killing. In addition, we further identified that the target of RocA was ULK1 (unc-51 like autophagy activating kinase 1) that is required for autophagy initiation. Using firefly luciferase containing the 5´ untranslated region of ULK1, we found that RocA inhibited the protein translation of ULK1 in a sequence-specific manner. Taken together, RocA could block autophagic immune resistance to NK cell-mediated killing, and our data suggested that RocA was a promising therapeutic candidate in NK cell-based cancer immunotherapy.
Collapse
Affiliation(s)
- Chao Yao
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Zhongya Ni
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Chenyuan Gong
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Xiaowen Zhu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Lixin Wang
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Zihang Xu
- Department of Internal Classic of Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Chunxian Zhou
- Department of Pathology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Suyun Li
- Department of Pathology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Wuxiong Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Chunpu Zou
- Department of Internal Classic of Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Shiguo Zhu
- Laboratory of Integrative Medicine, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
- Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
28
|
Djehal A, Krayem M, Najem A, Hammoud H, Cresteil T, Nebigil CG, Wang D, Yu P, Bentouhami E, Ghanem GE, Désaubry L. Targeting prohibitin with small molecules to promote melanogenesis and apoptosis in melanoma cells. Eur J Med Chem 2018; 155:880-888. [DOI: 10.1016/j.ejmech.2018.06.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/18/2018] [Accepted: 06/22/2018] [Indexed: 12/22/2022]
|
29
|
Significance of prohibitin domain family in tumorigenesis and its implication in cancer diagnosis and treatment. Cell Death Dis 2018; 9:580. [PMID: 29784973 PMCID: PMC5962566 DOI: 10.1038/s41419-018-0661-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Prohibitin (PHB) was originally isolated and characterized as an anti-proliferative gene in rat liver. The evolutionarily conserved PHB gene encodes two human protein isoforms with molecular weights of ~33 kDa, PHB1 and PHB2. PHB1 and PHB2 belong to the prohibitin domain family, and both are widely distributed in different cellular compartments such as the mitochondria, nucleus, and cell membrane. Most studies have confirmed differential expression of PHB1 and PHB2 in cancers compared to corresponding normal tissues. Furthermore, studies verified that PHB1 and PHB2 are involved in the biological processes of tumorigenesis, including cancer cell proliferation, apoptosis, and metastasis. Two small molecule inhibitors, Rocaglamide (RocA) and fluorizoline, derived from medicinal plants, were demonstrated to interact directly with PHB1 and thus inhibit the interaction of PHB with Raf-1, impeding Raf-1/ERK signaling cascades and significantly suppressing cancer cell metastasis. In addition, a short peptide ERAP and a natural product xanthohumol were shown to target PHB2 directly and prohibit cancer progression in estrogen-dependent cancers. As more efficient biomarkers and targets are urgently needed for cancer diagnosis and treatment, here we summarize the functional role of prohibitin domain family proteins, focusing on PHB1 and PHB2 in tumorigenesis and cancer development, with the expectation that targeting the prohibitin domain family will offer more clues for cancer therapy.
Collapse
|
30
|
Yuan G, Chen X, Liu Z, Wei W, Shu Q, Abou-Hamdan H, Jiang L, Li X, Chen R, Désaubry L, Zhou F, Xie D. Flavagline analog FL3 induces cell cycle arrest in urothelial carcinoma cell of the bladder by inhibiting the Akt/PHB interaction to activate the GADD45α pathway. J Exp Clin Cancer Res 2018; 37:21. [PMID: 29415747 PMCID: PMC5804081 DOI: 10.1186/s13046-018-0695-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Prohibitin 1 (PHB) is a potential target for the treatment of urothelial carcinoma of the bladder (UCB). FL3 is a newly synthesized agent that inhibits cancer cell proliferation by targeting the PHB protein; however, the effect of FL3 in UCB cells remains unexplored. METHODS FL3 was identified to be a potent inhibitor of UCB cell viability using CCK-8 (cell counting kit-8) assay. Then a series of in vitro and in vivo experiments were conducted to further demonstrate the inhibitory effect of FL3 on UCB cell proliferation and to determine the underlying mechanisms. RESULTS FL3 inhibited UCB cell proliferation and growth both in vitro and in vivo. By targeting the PHB protein, FL3 inhibited the interaction of Akt and PHB as well as Akt-mediated PHB phosphorylation, which consequently decreases the localization of PHB in the mitochondria. In addition, FL3 treatment resulted in cell cycle arrest in the G2/M phase, and this inhibitory effect of FL3 could be mimicked by knockdown of PHB. Through the microarray analysis of mRNA expression after FL3 treatment and knockdown of PHB, we found that the mRNA expression of the growth arrest and DNA damage-inducible alpha (GADD45α) gene were significantly upregulated. When knocked down the expression of GADD45α, the inhibitory effect of FL3 on cell cycle was rescued, suggesting that FL3-induced cell cycle inhibition is GADD45α dependent. CONCLUSION Our data provide that FL3 inhibits the interaction of Akt and PHB, which in turn activates the GADD45α-dependent cell cycle inhibition in the G2/M phase.
Collapse
Affiliation(s)
- Gangjun Yuan
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhuowei Liu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wensu Wei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qinghai Shu
- School of Material Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Hussein Abou-Hamdan
- Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg, Strasbourg, France
| | - Lijuan Jiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangdong Li
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rixin Chen
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Laurent Désaubry
- Therapeutic Innovation Laboratory, UMR7200, CNRS/University of Strasbourg, Strasbourg, France.
- Sino-French Joint Lab of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| | - Fangjian Zhou
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Dan Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|