1
|
Durán AM, Whitley K, Santiago K, Yoo C, Valdez G, Cheng KW, Ochoa P, de Semir D, Xiu J, Chokkalingam P, Das S, Schaefer ES, Rowe SP, Das BC, Casiano CA, Almaguel F. Inhibition of Mitochondrial-Associated Protein MAGMAS Resensitizes Chemoresistant Prostate Cancer Cells to Docetaxel. Cancers (Basel) 2025; 17:1535. [PMID: 40361461 PMCID: PMC12072152 DOI: 10.3390/cancers17091535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/15/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic prostate cancer (PCa) is the leading cause of cancer-related deaths and a major contributor to cancer mortality in men. Most patients with metastatic PCa eventually develop metastatic castration-resistant prostate cancer (mCRPC), characterized by resistance to treatment with androgen-deprivation therapy, and often later the development of resistance to other types of agents. MAGMAS, a 13.8 kDa mitochondrial-associated protein, facilitates the import of nuclear-encoded proteins into the mitochondrial matrix. Overexpression of MAGMAS has been observed in several aggressive cancers, including breast, glioblastoma, and prostate cancer. When overexpressed, MAGMAS acts as a cytoprotective protein by scavenging reactive oxygen species (ROS), maintaining ROS levels that support cell proliferation while avoiding the induction of apoptosis. This study investigates the role of MAGMAS in therapy resistance in PCa cells. METHODS/RESULTS Quantitative immunoblotting revealed that MAGMAS is endogenously upregulated in docetaxel-resistant (DR) PCa cell lines compared to their docetaxel-sensitive parental counterparts. While MAGMAS depletion alone did not affect the survival of DR cells, it significantly sensitized them to docetaxel (DTX), as indicated by a marked reduction in clonogenic potential. Additionally, transient knockdown of MAGMAS in these resistant cells significantly decreased the levels of ABCB1 protein. Consistent with these findings, sub-therapeutic inhibition of MAGMAS using the novel BT#9 inhibitor, in combination with increasing concentrations of DTX, enhanced the sensitivity of DR cells to DTX, as demonstrated by proliferation and clonogenic assays. Lastly, RNA tumor expression predicts overall survival (OS). CONCLUSIONS These results implicate MAGMAS in PCa chemoresistance and suggest that targeting this protein could provide a novel therapeutic strategy for treating DR tumors.
Collapse
Affiliation(s)
- Alfonso M. Durán
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Kristen Whitley
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Krystal Santiago
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Christian Yoo
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Giancarlo Valdez
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Kai Wen Cheng
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - Pedro Ochoa
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
| | - David de Semir
- Caris Life Sciences, Phoenix, AZ 85040, USA; (D.d.S.); (J.X.)
| | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ 85040, USA; (D.d.S.); (J.X.)
| | - Parthiban Chokkalingam
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | - Sasmita Das
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | | | - Steven P. Rowe
- Department of Radiology, University of North Carolina, Chapel Hill, NC 27514, USA;
| | - Bhaskar C. Das
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY 14215, USA; (P.C.); (S.D.); (B.C.D.)
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
- Department of Medicine, Rheumatology Division, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Frankis Almaguel
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; (A.M.D.); (K.S.); (C.Y.); (K.W.C.); (P.O.); (C.A.C.)
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
- Cancer Center, Loma Linda University Health, Loma Linda, CA 92354, USA
| |
Collapse
|
2
|
Raza A, Hoque A, Luwor R, Escalona RM, Kelly J, Sharma R, Charchar F, Chu S, Short MK, Jubinsky PT, Kannourakis G, Ahmed N. Enhanced Expression of Mitochondrial Magmas Protein in Ovarian Carcinomas: Magmas Inhibition Facilitates Antitumour Effects, Signifying a Novel Approach for Ovarian Cancer Treatment. Cells 2025; 14:655. [PMID: 40358179 PMCID: PMC12071367 DOI: 10.3390/cells14090655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/13/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondrial-associated granulocyte macrophage colony-stimulating factor (Magmas) is a unique protein located in the inner membrane of mitochondria, with an active role in scavenging reactive oxygen species (ROS) in cellular systems. Ovarian cancer (OC), one of the deadliest gynaecological cancers, is characterised by genomic instability, affected by ROS production in the tumour microenvironment. This manuscript discusses the role of Magmas and efficacy of its novel small molecule inhibitor BT#9 in OC progression, metastasis, and chemoresistance. Magmas expression levels were significantly elevated in high-grade human OC compared to benign tumours by immunohistochemistry. The inhibition of Magmas by BT#9 enhanced ROS production and reduced mitochondrial membrane permeability, basal respiration, mitochondrial ATP production, and cellular functions, such as the proliferation and migration of OC cell lines in vitro. Oral administration of BT#9 in vivo significantly reduced tumour growth and spread and enhanced the survival of mice without having any effect on the peritoneal organs. These data suggest that Magmas is functionally important for OC growth and spread by affecting ROS levels and that the inhibition of Magmas activity by BT#9 may provide novel clinical benefits for patients with this malignancy.
Collapse
Affiliation(s)
- Ali Raza
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
- Institute of Innovation, Science and Sustainability, Federation University Australia, Health Innovation and Transformation Center, Mount Helen Campus, Ballarat, VIC 3050, Australia;
| | - Ashfaqul Hoque
- St Vincent’s Institute of Medical Research, Fitzroy, VIC 3065, Australia;
| | - Rodney Luwor
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
- Institute of Innovation, Science and Sustainability, Federation University Australia, Health Innovation and Transformation Center, Mount Helen Campus, Ballarat, VIC 3050, Australia;
- Department of Surgery, Faculty of Medicine, Dentistry and Health Sciences, Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Ruth M. Escalona
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
- Centre for Endocrinology and Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular & Translational Science, Faculty of Medicine, Nursing and Health Sciences, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Jason Kelly
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
| | - Revati Sharma
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
| | - Fadi Charchar
- Institute of Innovation, Science and Sustainability, Federation University Australia, Health Innovation and Transformation Center, Mount Helen Campus, Ballarat, VIC 3050, Australia;
| | - Simon Chu
- Centre for Endocrinology and Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular & Translational Science, Faculty of Medicine, Nursing and Health Sciences, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Mary K. Short
- Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA; (M.K.S.); (P.T.J.)
| | - Paul T. Jubinsky
- Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA; (M.K.S.); (P.T.J.)
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
- Institute of Innovation, Science and Sustainability, Federation University Australia, Health Innovation and Transformation Center, Mount Helen Campus, Ballarat, VIC 3050, Australia;
| | - Nuzhat Ahmed
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3353, Australia; (A.R.); (R.L.); (R.M.E.); (J.K.); (R.S.); (G.K.)
- Institute of Innovation, Science and Sustainability, Federation University Australia, Health Innovation and Transformation Center, Mount Helen Campus, Ballarat, VIC 3050, Australia;
- Centre for Endocrinology and Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
- Department of Molecular & Translational Science, Faculty of Medicine, Nursing and Health Sciences, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Dentistry and Health Sciences, Royal Women’s Hospital, University of Melbourne, Parkville, VIC 3050, Australia
- Department of Surgery, Faculty of Medicine, Dentistry and Health Sciences, St Vincent Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
3
|
Motahari Z, Lepe JJ, Bautista MR, Hoerig C, Plant-Fox AS, Das B, Fowler CD, Magge SN, Bota DA. Preclinical assessment of MAGMAS inhibitor as a potential therapy for pediatric medulloblastoma. PLoS One 2024; 19:e0300411. [PMID: 39436961 PMCID: PMC11495579 DOI: 10.1371/journal.pone.0300411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/01/2024] [Indexed: 10/25/2024] Open
Abstract
Medulloblastoma is the most common malignant brain tumor in children. It has WNT-driven, SHH-driven/TP53 mutant, SHH-driven/TP53 wildtype, and non-WNT/non-SHH subgroups. MAGMAS (Mitochondrial Associated Granulocyte Macrophage colony-stimulating factor Signaling molecules) encodes a mitochondrial import inner membrane translocase subunit and is responsible for the translocation of matrix proteins across the inner membrane. We previously reported that a small molecule MAGMAS inhibitor, BT9, decreases cell proliferation, migration, and oxidative phosphorylation in adult glioblastoma cell lines. The aim of our study was to investigate whether the chemotherapeutic effect of BT9 can be extended to pediatric medulloblastoma. METHODS DAOY (SHH driven/tp53 mutant) and D425 (non-SHH group 3) were treated with BT9. For in vitro analysis, cell proliferation, death, migration, invasion, and metabolic activity were assessed using MTT assay, TUNEL staining, scratch wound assay, Matrigel invasion chambers, and seahorse assay, respectively. A D425 orthotopic xenograft mouse model was used to evaluate BT9 efficacy in vivo. RESULTS BT9 treatment resulted in a significant decrease in cell proliferation (DAOY, 24 hours IC50: 3.6 μM, 48 hours IC50: 2.3 μM, 72 hours IC50: 2.1 μM; D425 24 hours IC50: 3.4 μM, 48 hours IC50: 2.2 μM, 72 hours IC50: 2.1 μM) and a significant increase in cell death (DAOY, 24 hours p = 0.0004, 48 hours p<0.0001; D425, 24 hours p = 0.0001, 48 hours p = 0.02). In DAOY cells, 3 μM BT9 delayed migration and significantly reduced DAOY and D425 cell invasion (p < 0.0001). It also modified mitochondrial respiratory function in both medulloblastoma cell lines. Compared to control, however, BT9 administration did not improve survival in a D425 orthotopic xenograft mouse model. CONCLUSIONS Our in vitro data showed BT9 antitumor efficacy in DAOY and D425 cell lines, suggesting that BT9 may represent a promising targeted therapeutic in pediatric medulloblastoma. These data, however, need to be further validated in animal models.
Collapse
Affiliation(s)
- Zahra Motahari
- CHOC Neuroscience Institute, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Pediatrics, University of Irvine, Irvine, CA, United States of America
| | - Javier J. Lepe
- Department of Neurology, School of Medicine, University of Irvine, Irvine, CA, United States of America
| | - Malia R. Bautista
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA, United States of America
| | - Clay Hoerig
- Department of Pediatric Oncology, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States of America
| | - Ashley S. Plant-Fox
- Department of Pediatric Oncology, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Pediatric Oncology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL, United States of America
| | - Bhaskar Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, United States of America
- Department of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Christie D. Fowler
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA, United States of America
| | - Suresh N. Magge
- CHOC Neuroscience Institute, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Neurosurgery, Children’s Hospital of Orange County, Orange, CA, United States of America
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States of America
| | - Daniela A. Bota
- Department of Neurology, School of Medicine, University of Irvine, Irvine, CA, United States of America
| |
Collapse
|
4
|
Motahari Z, Lepe JJ, Bautista MR, Hoerig C, Plant-Fox AS, Das B, Fowler CD, Magge SN, Bota DA. Preclinical assessment of MAGMAS inhibitor as a potential therapy for pediatric medulloblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582709. [PMID: 38464047 PMCID: PMC10925277 DOI: 10.1101/2024.02.29.582709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Medulloblastoma, the most common pediatric brain malignancy, has Sonic Hedgehog (SHH) and non-SHH group3 subtypes. MAGMAS (Mitochondrial Associated Granulocyte Macrophage colony-stimulating factor Signaling molecules) encode for mitochondrial import inner membrane translocase subunit and is responsible for translocation of matrix proteins across the inner membrane. We previously reported that a small molecule MAGMAS inhibitor, BT9, decreases cell proliferation, migration, and oxidative phosphorylation in adult glioblastoma cell lines. The aim of our study was to investigate whether the chemotherapeutic effect of BT9 can be extended to pediatric medulloblastoma. Methods Multiple in vitro assays were performed using human DAOY (SHH activated tp53 mutant) and D425 (non-SHH group 3) cells. The impact of BT9 on cellular growth, death, migration, invasion, and metabolic activity were quantified using MTT assay, TUNEL staining, scratch wound assay, Matrigel invasion chambers, and seahorse assay, respectively. Survival following 50mg/kg BT9 treatment was assessed in vivo in immunodeficient mice intracranially implanted with D425 cells. Results Compared to control, BT9 treatment led to a significant reduction in medulloblastoma cell growth (DAOY, 24hrs IC50: 3.6uM, 48hrs IC50: 2.3uM, 72hrs IC50: 2.1uM; D425 24hrs IC50: 3.4uM, 48hrs IC50: 2.2uM, 72hrs IC50: 2.1uM) and a significant increase in cell death (DAOY, 24hrs p=0.0004, 48hrs p<0.0001; D425, 24hrs p=0.0001, 48hrs p=0.02). In DAOY cells, 3uM BT9 delayed migration, and significantly decreased DAOY and D425 cells invasion (p < 0.0001). Our in vivo study, however, did not extend survival in xenograft mouse model of group3 medulloblastoma compared to vehicle-treated controls. Conclusions Our in vitro data showed BT9 antitumor efficacy in DAOY and D425 cell lines suggesting that BT9 may represent a promising targeted therapeutic in pediatric medulloblastoma. These data, however, need to be further validated in animal models.
Collapse
Affiliation(s)
- Zahra Motahari
- CHOC Neuroscience Institute, Children's Hospital of Orange County, Orange, CA, USA
- Department of Pediatrics, University of Irvine, CA, USA
| | - Javier J Lepe
- Department of Neurology, School of Medicine, University of Irvine, CA, USA
| | - Malia R Bautista
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Clay Hoerig
- Department of Pediatric Oncology, Children's Hospital of Orange County, Orange, CA, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Ashley S Plant-Fox
- Department of Pediatric Oncology, Children's Hospital of Orange County, Orange, CA, USA
- Department of Pediatric Oncology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Bhaskar Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
- Department of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christie D Fowler
- Department of Neurobiology and Behavior, School of Biological Sciences, University of California, Irvine, CA, USA
| | - Suresh N Magge
- CHOC Neuroscience Institute, Children's Hospital of Orange County, Orange, CA, USA
- Department of Neurosurgery, Children's Hospital of Orange County, Orange, CA, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Daniela A Bota
- Department of Neurology, School of Medicine, University of Irvine, CA, USA
| |
Collapse
|
5
|
Lazarus HM, Pitts K, Wang T, Lee E, Buchbinder E, Dougan M, Armstrong DG, Paine R, Ragsdale CE, Boyd T, Rock EP, Gale RP. Recombinant GM-CSF for diseases of GM-CSF insufficiency: Correcting dysfunctional mononuclear phagocyte disorders. Front Immunol 2023; 13:1069444. [PMID: 36685591 PMCID: PMC9850113 DOI: 10.3389/fimmu.2022.1069444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction Endogenous granulocyte-macrophage colony-stimulating factor (GM-CSF), identified by its ability to support differentiation of hematopoietic cells into several types of myeloid cells, is now known to support maturation and maintain the metabolic capacity of mononuclear phagocytes including monocytes, macrophages, and dendritic cells. These cells sense and attack potential pathogens, present antigens to adaptive immune cells, and recruit other immune cells. Recombinant human (rhu) GM-CSF (e.g., sargramostim [glycosylated, yeast-derived rhu GM-CSF]) has immune modulating properties and can restore the normal function of mononuclear phagocytes rendered dysfunctional by deficient or insufficient endogenous GM-CSF. Methods We reviewed the emerging biologic and cellular effects of GM-CSF. Experts in clinical disease areas caused by deficient or insufficient endogenous GM-CSF examined the role of GM-CSF in mononuclear phagocyte disorders including autoimmune pulmonary alveolar proteinosis (aPAP), diverse infections (including COVID-19), wound healing, and anti-cancer immune checkpoint inhibitor therapy. Results We discuss emerging data for GM-CSF biology including the positive effects on mitochondrial function and cell metabolism, augmentation of phagocytosis and efferocytosis, and immune cell modulation. We further address how giving exogenous rhu GM-CSF may control or treat mononuclear phagocyte dysfunction disorders caused or exacerbated by GM-CSF deficiency or insufficiency. We discuss how rhu GM-CSF may augment the anti-cancer effects of immune checkpoint inhibitor immunotherapy as well as ameliorate immune-related adverse events. Discussion We identify research gaps, opportunities, and the concept that rhu GM-CSF, by supporting and restoring the metabolic capacity and function of mononuclear phagocytes, can have significant therapeutic effects. rhu GM-CSF (e.g., sargramostim) might ameliorate multiple diseases of GM-CSF deficiency or insufficiency and address a high unmet medical need.
Collapse
Affiliation(s)
- Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, United States
| | - Katherine Pitts
- Medical Affairs, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Tisha Wang
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elinor Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Elizabeth Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - David G. Armstrong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Robert Paine
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT, United States
| | | | - Timothy Boyd
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Edwin P. Rock
- Clinical Development, Partner Therapeutics, Inc., Lexington, MA, United States
| | - Robert Peter Gale
- Hematology Centre, Department of Immunology and Inflammation, Imperial College, London, United Kingdom
| |
Collapse
|
6
|
Yang J, Das BC, Aljitawi O, Kumar A, Das S, Van Veldhuizen P. Magmas Inhibition in Prostate Cancer: A Novel Target for Treatment-Resistant Disease. Cancers (Basel) 2022; 14:cancers14112732. [PMID: 35681713 PMCID: PMC9179500 DOI: 10.3390/cancers14112732] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 01/25/2023] Open
Abstract
Simple Summary Metastatic and treatment-resistant prostate cancer remains a life-threatening disease despite recent therapeutic advances. Literature suggests that treatment resistance and prostate cancer progression is associated with prostate cancer stem cells. In this study, we evaluated the role of the mitochondria-associated granulocyte–macrophage colony-stimulating factor signaling (Magmas) protein as a molecular target and applied a novel Magmas inhibitor, BT#9, on prostate cancer cells and normal control cells. We found that Magmas was overexpressed in human prostate cancers and its expression was linked to the aggressiveness of the disease. BT#9 downregulated Magmas expression, reduced viability and induced apoptotic cell death in prostate cancer cells. The mechanism of cell death by BT#9 is mainly caspase-independent and via a Reactive Oxygen Species (ROS)-mediated pathway. This is the first study that has evaluated targeting the Magmas protein in prostate cancer and, to our knowledge, the first to elucidate the potential molecular mechanism of BT#9 activity in prostate cancer, including the mode of cell death and the critical role of ROS accumulation. Our work may provide a potential clinical application for a novel prostate cancer treatment that can overcome cancer stem cell and therapeutic resistance. Abstract The purpose of our study was to evaluate Magmas as a potential target in prostate cancer. In addition, we evaluated our synthetic Magmas inhibitor (BT#9) effects on prostate cancer and examined the molecular mechanism of BT#9. A cell viability assay showed that treatment with BT#9 caused a significant decrease in the viability of DU145 and PC3 prostate cancer cells with little effect on the viability of WPMY-1 normal prostate cells. Western blot proved that BT#9 downregulated the Magmas protein and caspase-3 activation. Flow cytometry studies demonstrated increased apoptosis and disturbed mitochondrial membrane potential. However, the main mode of cell death was caspase-independent necrosis, which was correlated with the accumulation of mitochondrial and intra-cellular Reactive Oxygen Species (ROS). Taken together, our data suggest Magmas is a potential molecular target for the treatment of prostate cancer and that Magmas inhibition results in ROS-dependent and caspase-independent necrotic cell death.
Collapse
Affiliation(s)
- Jianhui Yang
- Wilmot Cancer Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (O.A.)
| | - Bhaskar C. Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.K.); (S.D.)
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: (B.C.D.); (P.V.V.)
| | - Omar Aljitawi
- Wilmot Cancer Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (O.A.)
| | - Avinash Kumar
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.K.); (S.D.)
| | - Sasmita Das
- Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA; (A.K.); (S.D.)
| | - Peter Van Veldhuizen
- Wilmot Cancer Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; (J.Y.); (O.A.)
- Correspondence: (B.C.D.); (P.V.V.)
| |
Collapse
|
7
|
Waingankar TP, D'Silva P. Multiple variants of the human presequence translocase motor subunit Magmas govern the mitochondrial import. J Biol Chem 2021; 297:101349. [PMID: 34715125 PMCID: PMC8605242 DOI: 10.1016/j.jbc.2021.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 11/22/2022] Open
Abstract
Mitochondrial protein translocation is an intricately regulated process that requires dedicated translocases at the outer and inner membranes. The presequence translocase complex, translocase of the inner membrane 23, facilitates most of the import of preproteins containing presequences into the mitochondria, and its primary structural organization is highly conserved. As part of the translocase motor, two J-proteins, DnaJC15 and DnaJC19, are recruited to form two independent translocation machineries (translocase A and translocase B, respectively). On the other hand, the J-like protein subunit of translocase of the inner membrane 23, Mitochondria-associated granulocyte-macrophage colony-stimulating factor signaling molecule (Magmas) (orthologous to the yeast subunit Pam16), can regulate human import-motor activity by forming a heterodimer with DnaJC19 and DnaJC15. However, the precise coordinated regulation of two human import motors by a single Magmas protein is poorly understood. Here, we report two additional Magmas variants (Magmas-1 and Magmas-2) constitutively expressed in the mammalian system. Both the Magmas variants are functional orthologs of Pam16 with an evolutionarily conserved J-like domain critical for cell survival. Moreover, the Magmas variants are peripherally associated with the inner membrane as part of the human import motor for translocation. Our results demonstrate that Magmas-1 is predominantly recruited to translocase B, whereas Magmas-2 is majorly associated with translocase A. Strikingly, both the variants exhibit differential J-protein inhibitory activity in modulating import motor, thereby regulating overall translocase function. Based on our findings, we hypothesize that additional Magmas variants are of evolutionary significance in humans to maximize protein import in familial-linked pathological conditions.
Collapse
Affiliation(s)
| | - Patrick D'Silva
- Department of Biochemistry, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
8
|
Palmer CS, Anderson AJ, Stojanovski D. Mitochondrial protein import dysfunction: mitochondrial disease, neurodegenerative disease and cancer. FEBS Lett 2021; 595:1107-1131. [PMID: 33314127 DOI: 10.1002/1873-3468.14022] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/12/2020] [Accepted: 10/17/2020] [Indexed: 12/13/2022]
Abstract
The majority of proteins localised to mitochondria are encoded by the nuclear genome, with approximately 1500 proteins imported into mammalian mitochondria. Dysfunction in this fundamental cellular process is linked to a variety of pathologies including neuropathies, cardiovascular disorders, myopathies, neurodegenerative diseases and cancer, demonstrating the importance of mitochondrial protein import machinery for cellular function. Correct import of proteins into mitochondria requires the co-ordinated activity of multimeric protein translocation and sorting machineries located in both the outer and inner mitochondrial membranes, directing the imported proteins to the destined mitochondrial compartment. This dynamic process maintains cellular homeostasis, and its dysregulation significantly affects cellular signalling pathways and metabolism. This review summarises current knowledge of the mammalian mitochondrial import machinery and the pathological consequences of mutation of its components. In addition, we will discuss the role of mitochondrial import in cancer, and our current understanding of the role of mitochondrial import in neurodegenerative diseases including Alzheimer's disease, Huntington's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Catherine S Palmer
- Department of Biochemistry and Molecular Biology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| | - Alexander J Anderson
- Department of Biochemistry and Molecular Biology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| | - Diana Stojanovski
- Department of Biochemistry and Molecular Biology and The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Australia
| |
Collapse
|
9
|
Ovarian Cancer, Cancer Stem Cells and Current Treatment Strategies: A Potential Role of Magmas in the Current Treatment Methods. Cells 2020; 9:cells9030719. [PMID: 32183385 PMCID: PMC7140629 DOI: 10.3390/cells9030719] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer (EOC) constitutes 90% of ovarian cancers (OC) and is the eighth most common cause of cancer-related death in women. The cancer histologically and genetically is very complex having a high degree of tumour heterogeneity. The pathogenic variability in OC causes significant impediments in effectively treating patients, resulting in a dismal prognosis. Disease progression is predominantly influenced by the peritoneal tumour microenvironment rather than properties of the tumor and is the major contributor to prognosis. Standard treatment of OC patients consists of debulking surgery, followed by chemotherapy, which in most cases end in recurrent chemoresistant disease. This review discusses the different origins of high-grade serous ovarian cancer (HGSOC), the major sub-type of EOC. Tumour heterogeneity, genetic/epigenetic changes, and cancer stem cells (CSC) in facilitating HGSOC progression and their contribution in the circumvention of therapy treatments are included. Several new treatment strategies are discussed including our preliminary proof of concept study describing the role of mitochondria-associated granulocyte macrophage colony-stimulating factor signaling protein (Magmas) in HGSOC and its unique potential role in chemotherapy-resistant disease.
Collapse
|
10
|
Li N, Zhan X. Mitochondrial Dysfunction Pathway Networks and Mitochondrial Dynamics in the Pathogenesis of Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:690. [PMID: 31649621 PMCID: PMC6794370 DOI: 10.3389/fendo.2019.00690] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondrion is a multi-functional organelle, which is associated with various signaling pathway networks, including energy metabolism, oxidative stress, cell apoptosis, cell cycles, autophagy, and immunity process. Mitochondrial proteins have been discovered to modulate these signaling pathway networks, and multiple biological behaviors to adapt to various internal environments or signaling events of human pathogenesis. Accordingly, mitochondrial dysfunction that alters the bioenergetic and biosynthetic state might contribute to multiple diseases, including cell transformation and tumor. Multiomics studies have revealed that mitochondrial dysfunction, oxidative stress, and cell cycle dysregulation signaling pathways operate in human pituitary adenomas, which suggest mitochondria play critical roles in pituitary adenomas. Some drugs targeting mitochondria are found as a therapeutic strategy for pituitary adenomas, including melatonin, melatonin inhibitors, temozolomide, pyrimethamine, 18 beta-glycyrrhetinic acid, gossypol acetate, Yougui pill, T-2 toxin, grifolic acid, cyclosporine A, dopamine agonists, and paeoniflorin. This article reviews the latest experimental evidence and potential biological roles of mitochondrial dysfunction and mitochondrial dynamics in pituitary adenoma progression, potential molecular mechanisms between mitochondria and pituitary adenoma progression, and current status and perspectives of mitochondria-based biomarkers and targeted drugs for effective management of pituitary adenomas.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Di K, Lomeli N, Bota DA, Das BC. Magmas inhibition as a potential treatment strategy in malignant glioma. J Neurooncol 2018; 141:267-276. [PMID: 30414099 DOI: 10.1007/s11060-018-03040-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/29/2018] [Indexed: 11/30/2022]
Abstract
PURPOSE Magmas (mitochondria-associated protein involved in granulocyte-macrophage colony-stimulating factor signal transduction) is a nuclear gene that encodes the mitochondrial import inner membrane translocase subunit Tim16. Magmas is highly conserved, ubiquitously expressed in mammalian cells, and is essential for cell viability. Magmas expression levels are increased in prostate cancers and pituitary adenomas. Moreover, silencing Magmas by RNAi sensitizes pituitary adenoma cells to pro-apoptotic stimuli and induces a G0/G1 accumulation. The aim of this study was to examine whether inhibition of Magmas by small molecule inhibitors could be beneficial for the treatment of malignant gliomas. METHODS We evaluated the expression of Magmas in patient-derived glioblastoma tissue samples and xenograft models. We studied the feasibility of a small molecule Magmas inhibitor (BT#9) as a therapeutic agent in stable human glioma cell lines and high-grade patient derived glioma stem-like cells. RESULTS Magmas was overexpressed in tissue sections from glioma patients and xenografts. In vivo studies revealed that BT#9 could cross the blood-brain barrier in the animal model. Magmas inhibition by BT#9 in glioma cell lines significantly decreased cell proliferation, induced apoptosis along with vacuole formation, and blocked migration and invasion. In addition, BT#9 treatment decreased the respiratory function of glioma cells, supporting the role that Magmas serves as a reactive oxygen species regulator. CONCLUSIONS This is the first study on the role of Magmas in glioma. Our findings suggest that Magmas plays a key role in glioma cell survival and targeting Magmas by small molecule inhibitors may be a therapeutic strategy in gliomas.
Collapse
Affiliation(s)
- Kaijun Di
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Naomi Lomeli
- Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Daniela A Bota
- Department of Neurology, University of California Irvine, Irvine, CA, USA. .,Department of Pathology & Laboratory Medicine, University of California Irvine, Irvine, CA, USA. .,Department of Neurological Surgery, University of California Irvine, Irvine, CA, USA. .,Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA. .,, 200 S. Manchester Ave., Suite 206, Orange, CA, 92868, USA.
| | - Bhaskar C Das
- Department of Medicine and Pharmacological Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
12
|
Gentilin E, Minoia M, Bondanelli M, Tagliati F, Degli Uberti EC, Zatelli MC. Growth Hormone differentially modulates chemoresistance in human endometrial adenocarcinoma cell lines. Endocrine 2017; 56:621-632. [PMID: 27585662 DOI: 10.1007/s12020-016-1085-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023]
Abstract
Growth Hormone may influence neoplastic development of endometrial epithelium towards endometrial adenocarcinoma, which is one of the most occurring tumors in acromegalic patients. Since chemoresistance often develops in advanced endometrial adenocarcinoma, we investigated whether Growth Hormone might influence the development of chemoresistance to drugs routinely employed in endometrial adenocarcinoma treatment, such as Doxorubicin, Cisplatin, and Paclitaxel. Growth Hormone and Growth Hormone receptor expression was assessed by immunofluorescence in two endometrial adenocarcinoma cell lines, AN3 CA and HEC-1-A cells. Growth Hormone effects were assessed investigating cell viability, caspase3/7 activation, ERK1/2, and protein kinase C delta protein expression. AN3 CA and HEC-1-A cells display Growth Hormone and Growth Hormone receptor. Growth Hormone does not influence cell viability in both cells lines, but significantly reduces caspase 3/7 activation in AN3 CA cells, an effect blocked by a Growth Hormone receptor antagonist. Growth Hormone rescues AN3 CA cells from the inhibitory effects of Doxorubicin and Cisplatin on cell viability, while it has no effect on Paclitaxel. Growth Hormone does not influence the pro-apoptotic effects of Doxorubicin, but is capable of rescuing AN3 CA cells from the pro-apoptotic effects of Cisplatin. On the other hand, Growth Hormone did not influence the effects of Doxorubicin and Paclitaxel on HEC-1A cell viability. The protective action of Growth Hormone towards the effects of Doxorubicin may be mediated by ERK1/2 activation, while the pro-apoptotic effects of Cisplatin may be mediated by protein kinase C delta inhibition. All together our results indicate that Growth Hormone may differentially contribute to endometrial adenocarcinoma chemoresistance. This may provide new insights on novel therapies against endometrial adenocarcinoma chemoresistant aggressive tumors.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mariella Minoia
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Marta Bondanelli
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Federico Tagliati
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Ettore C Degli Uberti
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy
- Laboratorio in rete del Tecnopolo "Tecnologie delle terapie avanzate" (LTTA) of the University of Ferrara, Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Ferrara, Italy.
- Laboratorio in rete del Tecnopolo "Tecnologie delle terapie avanzate" (LTTA) of the University of Ferrara, Ferrara, Italy.
| |
Collapse
|
13
|
Miyata N, Tang Z, Conti MA, Johnson ME, Douglas CJ, Hasson SA, Damoiseaux R, Chang CEA, Koehler CM. Adaptation of a Genetic Screen Reveals an Inhibitor for Mitochondrial Protein Import Component Tim44. J Biol Chem 2017; 292:5429-5442. [PMID: 28167535 DOI: 10.1074/jbc.m116.770131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/31/2017] [Indexed: 12/28/2022] Open
Abstract
Diverse protein import pathways into mitochondria use translocons on the outer membrane (TOM) and inner membrane (TIM). We adapted a genetic screen, based on Ura3 mistargeting from mitochondria to the cytosol, to identify small molecules that attenuated protein import. Small molecule mitochondrial import blockers of the Carla Koehler laboratory (MB)-10 inhibited import of substrates that require the TIM23 translocon. Mutational analysis coupled with molecular docking and molecular dynamics modeling revealed that MB-10 binds to a specific pocket in the C-terminal domain of Tim44 of the protein-associated motor (PAM) complex. This region was proposed to anchor Tim44 to the membrane, but biochemical studies with MB-10 show that this region is required for binding to the translocating precursor and binding to mtHsp70 in low ATP conditions. This study also supports a direct role for the PAM complex in the import of substrates that are laterally sorted to the inner membrane, as well as the mitochondrial matrix. Thus, MB-10 is the first small molecule modulator to attenuate PAM complex activity, likely through binding to the C-terminal region of Tim44.
Collapse
Affiliation(s)
- Non Miyata
- From the Departments of Chemistry and Biochemistry and
| | - Zhiye Tang
- the Department of Chemistry, University of California Riverside, Riverside, California 92521
| | | | | | | | | | | | - Chia-En A Chang
- the Department of Chemistry, University of California Riverside, Riverside, California 92521
| | - Carla M Koehler
- From the Departments of Chemistry and Biochemistry and .,the Molecular Biology Institute, and.,the Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California 90095 and
| |
Collapse
|
14
|
Abstract
MicroRNAs (miRNAs) are non-coding RNAs generated from endogenous hairpin-shaped transcripts that powerfully regulate gene expression at post-transcriptional level. Each miRNA is capable to regulate the expression levels of hundreds of transcripts and each mRNA may have more than one miRNA recognition sequence. There is emerging evidence that deregulation of miRNA expression leads to the alteration of pivotal physiological functions contributing to the development of diseases and neoplasms, including pituitary adenoma. This review is aimed at providing the up-to-date knowledge concerning deregulated miRNAs of pituitary tumors and their functions. In order to take stock, pituitary tumors have been sub-divided in different classes on the basis of tumor features (histotype, dimension, aggressiveness). The overview takes full consideration of the recent advances in miRNAs role as potential therapeutics and biomarkers.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Ettore Degli Uberti
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology and Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy.
| |
Collapse
|
15
|
Gentilin E, Di Pasquale C, Gagliano T, Tagliati F, Benfini K, Ambrosio MR, Bondanelli M, degli Uberti EC, Zatelli MC. Protein Kinase C Delta restrains growth in ACTH-secreting pituitary adenoma cells. Mol Cell Endocrinol 2016; 419:252-8. [PMID: 26522132 DOI: 10.1016/j.mce.2015.10.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/21/2015] [Accepted: 10/26/2015] [Indexed: 12/25/2022]
Abstract
Protein Kinase C Delta (PRKCD) has been highlighted among disrupted pathways in corticotroph adenomas. PRKCD is expressed at low level in human corticotroph adenomas and controls cell cycle in vitro. Therefore, PRKCD may play an important role in the development/progression of corticotroph adenomas, warranting further studies to understand the role of PRKCD and related pathways in restraining pituitary cell growth. We evaluated PRKCD role in influencing cell behavior in terms of cell viability, hormone expression and protein expression profile, by silencing PRKCD in AtT-20/D16v-F2 cells. PRKCD silencing increases cell viability, enhances hormone expression and induces morphological changes associated with deregulation of adhesion molecules. PRKCD silencing is associated with an increase in Epithelial Growth Factor Receptor (EGFR) expression, a marker of tumor aggressive behavior, and sensitivity to anti-EGFR molecules. PRKCD might restrain corticotroph adenoma cells from acquiring an aggressive behavior, candidating PRKCD as a possible molecular target for the treatment of corticotroph adenomas.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy; Laboratorio in rete del Tecnopolo "Tecnologie delle terapie avanzate" (LTTA) of the University of Ferrara, Italy
| | - Carmelina Di Pasquale
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Teresa Gagliano
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Federico Tagliati
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Katiuscia Benfini
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Maria Rosaria Ambrosio
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Marta Bondanelli
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy
| | - Ettore C degli Uberti
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy; Laboratorio in rete del Tecnopolo "Tecnologie delle terapie avanzate" (LTTA) of the University of Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology & Internal Medicine, Dept. of Medical Sciences, University of Ferrara, Italy; Laboratorio in rete del Tecnopolo "Tecnologie delle terapie avanzate" (LTTA) of the University of Ferrara, Italy.
| |
Collapse
|
16
|
Seltzer J, Ashton CE, Scotton TC, Pangal D, Carmichael JD, Zada G. Gene and protein expression in pituitary corticotroph adenomas: a systematic review of the literature. Neurosurg Focus 2015; 38:E17. [PMID: 25639319 DOI: 10.3171/2014.10.focus14683] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECT Functional corticotroph pituitary adenomas (PAs) secrete adrenocorticotropic hormone (ACTH) and are the cause of Cushing's disease, which accounts for 70% of all cases of Cushing's syndrome. Current classification systems for PAs rely primarily on laboratory hormone findings, tumor size and morphology, invasiveness, and immunohistochemical findings. Likewise, drug development for functional ACTH-secreting PAs (ACTH-PAs) is limited and has focused largely on blocking the production or downstream effects of excess cortisol. The authors aimed to summarize the findings from previous studies that explored gene and protein expression of ACTH-PAs to prioritize potential genetic and protein targets for improved molecular diagnosis and treatment of Cushing's disease. METHODS A systematic literature review was performed using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A PubMed search of select medical subject heading (MeSH) terms was performed to identify all studies that reported gene- and protein-expression findings in ACTH-PAs from January 1, 1990, to August 24, 2014, the day the search was performed. The inclusion criteria were studies on functional ACTH-PAs compared with normal pituitary glands, on human PA tissue only, with any method of analysis, and published in the English language. Studies using anything other than resected PA tissue, those that compared other adenoma types, those without baseline expression data, or those in which any pretreatment was delivered before analysis were excluded. RESULTS The primary search returned 1371 abstracts, of which 307 were found to be relevant. Of those, 178 were selected for secondary full-text analysis. Of these, 64 articles met the inclusion criteria and an additional 4 studies were identified from outside the search for a total of 68 included studies. Compared with the normal pituitary gland, significant gene overexpression in 43 genes and 22 proteins was reported, and gene underexpression in 58 genes and 15 proteins was reported. Immunohistochemistry was used in 39 of the studies, and reverse transcriptase polymerase chain reaction was used in 26 of the studies, primarily, and as validation for 4 others. Thirteen studies used both immunohistochemistry and reverse transcriptase polymerase chain reaction. Other methods used included microarray, in situ hybridization, Northern blot analysis, and Western blot analysis. Expression of prioritized genes emphasized in multiple studies were often validated on both the gene and protein levels. Genes/proteins found to be overexpressed in ACTH-PAs relative to the normal pituitary gland included hPTTG1/securin, NEUROD1/NeuroD1 (Beta2), HSD11B2/11β-hydroxysteroid dehydrogenase 2, AKT/Akt, protein kinase B, and CCND1/cyclin D1. Candidate genes/proteins found to be underexpressed in ACTH-PAs relative to the normal pituitary gland included CDKN1B/p27(Kip1), CDKN2A/p16, KISS1/kisspeptin, ACTHR/ACTH-R, and miR-493. CONCLUSIONS On the basis of the authors' systematic review, many significant gene and protein targets that may contribute to tumorigenesis, invasion, and hormone production/secretion of ACTH have been identified and validated in ACTH-PAs. Many of these potential targets have not been fully analyzed for their therapeutic and diagnostic potential but may represent candidate molecular targets for biomarker development and drug targeting. This review may help catalyze additional research efforts using modern profiling and sequencing techniques and alteration of gene expression.
Collapse
|
17
|
Sambugaro S, Di Ruvo M, Ambrosio MR, Pellegata NS, Bellio M, Guerra A, Buratto M, Foschini MP, Tagliati F, degli Uberti E, Zatelli MC. Early onset acromegaly associated with a novel deletion in CDKN1B 5'UTR region. Endocrine 2015; 49:58-64. [PMID: 25645465 DOI: 10.1007/s12020-015-0540-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/23/2015] [Indexed: 10/24/2022]
Abstract
Genetic alterations frequently are involved in the development of a pituitary adenoma in young age. We here characterize the functional role of a deletion in CDKN1B 5'-UTR region (c.-29_-26delAGAG) identified in an acromegalic patient that developed a growth hormone in pituitary adenoma during childhood. Our results show that the identified novel heterozygous deletion in the CDKN1B 5'-UTR region associates with a reduction in CDKN1B mRNA levels, a predicted altered secondary mRNA structure, and a reduced CDKN1B 5'-UTR transcriptional activity in vitro. The patient displayed loss of heterozygosity in the same CDKN1B 5'-UTR region at tissue level and the 5'UTR region containing the deleted sequence encompasses a GRE. These findings indicate that the identification of functional alterations of newly discovered genetic derangements need to be fully characterized and always correlated with the clinical manifestations.
Collapse
Affiliation(s)
- Silvia Sambugaro
- Section of Endocrinology and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zatelli MC, Gagliano T, Pelà M, Bianco S, Bertolasi V, Tagliati F, Guerrini R, degli Uberti E, Salvadori S, Trapella C. N-Carbamidoyl-4-((3-ethyl-2,4,4-trimethylcyclohexyl)methyl)benzamide Enhances Staurosporine Cytotoxic Effects Likely Inhibiting the Protective Action of Magmas toward Cell Apoptosis. J Med Chem 2014; 57:4606-14. [DOI: 10.1021/jm5000535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Maria Chiara Zatelli
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie
Avanzate (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Teresa Gagliano
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Michela Pelà
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Sara Bianco
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Valerio Bertolasi
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Federico Tagliati
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Remo Guerrini
- Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie
Avanzate (LTTA), University of Ferrara, 44121 Ferrara, Italy
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Ettore degli Uberti
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie
Avanzate (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Severo Salvadori
- Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie
Avanzate (LTTA), University of Ferrara, 44121 Ferrara, Italy
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| | - Claudio Trapella
- Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie
Avanzate (LTTA), University of Ferrara, 44121 Ferrara, Italy
- Department of Chemical and Pharmaceutical
Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy
| |
Collapse
|
19
|
Unraveling the intricate organization of mammalian mitochondrial presequence translocases: existence of multiple translocases for maintenance of mitochondrial function. Mol Cell Biol 2014; 34:1757-75. [PMID: 24636990 DOI: 10.1128/mcb.01527-13] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are indispensable organelles implicated in multiple aspects of cellular processes, including tumorigenesis. Heat shock proteins play a critical regulatory role in accurately delivering the nucleus-encoded proteins through membrane-bound presequence translocase (Tim23 complex) machinery. Although altered expression of mammalian presequence translocase components had been previously associated with malignant phenotypes, the overall organization of Tim23 complexes is still unsolved. In this report, we show the existence of three distinct Tim23 complexes, namely, B1, B2, and A, involved in the maintenance of normal mitochondrial function. Our data highlight the importance of Magmas as a regulator of translocase function and in dynamically recruiting the J-proteins DnaJC19 and DnaJC15 to individual translocases. The basic housekeeping function involves translocases B1 and B2 composed of Tim17b isoforms along with DnaJC19, whereas translocase A is nonessential and has a central role in oncogenesis. Translocase B, having a normal import rate, is essential for constitutive mitochondrial functions such as maintenance of electron transport chain complex activity, organellar morphology, iron-sulfur cluster protein biogenesis, and mitochondrial DNA. In contrast, translocase A, though dispensable for housekeeping functions with a comparatively lower import rate, plays a specific role in translocating oncoproteins lacking presequence, leading to reprogrammed mitochondrial functions and hence establishing a possible link between the TIM23 complex and tumorigenicity.
Collapse
|
20
|
Gentilin E, Molè D, Gagliano T, Minoia M, Ambrosio MR, Degli Uberti EC, Zatelli MC. Inhibitory effects of mitotane on viability and secretory activity in mouse gonadotroph cell lines. Reprod Toxicol 2014; 45:71-6. [PMID: 24486453 DOI: 10.1016/j.reprotox.2014.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/09/2014] [Accepted: 01/20/2014] [Indexed: 01/03/2023]
Abstract
Mitotane represents the mainstay medical treatment for metastatic, inoperable or recurrent adrenocortical carcinoma. Besides the well-known adverse events, mitotane therapy is associated also with endocrinological effects, including sexual and reproductive dysfunction. The majority of male patients undergoing adjuvant mitotane therapy show a picture of hypogonadism, characterized by low free testosterone and high sex hormone binding globulin levels and unmodified LH concentrations. Since mitotane has been shown to have direct pituitary effects, we investigated whether mitotane may influence both cell viability and function of gonadotroph cells in the settings of two pituitary cell lines. We found that mitotane reduces cell viability, induces apoptosis, modifies cell cycle phase distribution and secretion of gonadotroph cells. The present data strengthen previous evidence showing a direct mitotane effect at pituitary level and represent a possible explanation of the lack of LH increase following decrease in free testosterone in patients undergoing adjuvant mitotane therapy.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy
| | - Daniela Molè
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | - Teresa Gagliano
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | - Mariella Minoia
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy
| | | | - Ettore C Degli Uberti
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Italy; Laboratorio in Rete del Tecnopolo Tecnologie delle Terapie Avanzate (LTTA), University of Ferrara, Italy.
| |
Collapse
|
21
|
Tagliati F, Gagliano T, Gentilin E, Minoia M, Molè D, delgi Uberti EC, Zatelli MC. Magmas overexpression inhibits staurosporine induced apoptosis in rat pituitary adenoma cell lines. PLoS One 2013; 8:e75194. [PMID: 24069394 PMCID: PMC3775776 DOI: 10.1371/journal.pone.0075194] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 08/14/2013] [Indexed: 12/22/2022] Open
Abstract
Magmas is a nuclear gene that encodes for the mitochondrial import inner membrane translocase subunit Tim16. Magmas is overexpressed in the majority of human pituitary adenomas and in a mouse ACTH-secreting pituitary adenoma cell line. Here we report that Magmas is highly expressed in two out of four rat pituitary adenoma cell lines and its expression levels inversely correlate to the extent of cellular response to staurosporine in terms of apoptosis activation and cell viability. Magmas over-expression in rat GH/PRL-secreting pituitary adenoma GH4C1 cells leads to an increase in cell viability and to a reduction in staurosporine-induced apoptosis and DNA fragmentation, in parallel with the increase in Magmas protein expression. These results indicate that Magmas plays a pivotal role in response to pro-apoptotic stimuli and confirm and extend the finding that Magmas protects pituitary cells from staurosporine-induced apoptosis, suggesting its possible involvement in pituitary adenoma development.
Collapse
Affiliation(s)
- Federico Tagliati
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Teresa Gagliano
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Erica Gentilin
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Laboratorio in rete del Tecnopolo “Tecnologie delle terapie avanzate” (LTTA) of the University of Ferrara, Ferrara, Italy
| | - Mariella Minoia
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Daniela Molè
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Ettore C. delgi Uberti
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Laboratorio in rete del Tecnopolo “Tecnologie delle terapie avanzate” (LTTA) of the University of Ferrara, Ferrara, Italy
| | - Maria Chiara Zatelli
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Laboratorio in rete del Tecnopolo “Tecnologie delle terapie avanzate” (LTTA) of the University of Ferrara, Ferrara, Italy
- * E-mail:
| |
Collapse
|
22
|
Gentilin E, Tagliati F, Terzolo M, Zoli M, Lapparelli M, Minoia M, Ambrosio MR, Degli Uberti EC, Zatelli MC. Mitotane reduces human and mouse ACTH-secreting pituitary cell viability and function. J Endocrinol 2013; 218:275-85. [PMID: 23814013 DOI: 10.1530/joe-13-0210] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Medical therapy for Cushing's disease (CD) is currently based on agents mainly targeting adrenocortical function. Lately, pituitary-directed drugs have been developed, with limited efficacy. Mitotane, a potent adrenolytic drug, has been recently investigated for the treatment of CD, but the direct pituitary effects have not been clarified so far. The aim of our study was to investigate whether mitotane may affect corticotroph function and cell survival in the mouse pituitary cell line AtT20/D16v-F2 and in the primary cultures of human ACTH-secreting pituitary adenomas, as an in vitro model of pituitary corticotrophs. We found that in the AtT20/D16v-F2 cell line and in primary cultures, mitotane reduces cell viability by inducing caspase-mediated apoptosis and reduces ACTH secretion. In the AtT20/D16v-F2 cell line, mitotane reduces Pomc expression and blocks the stimulatory effects of corticotropin-releasing hormone on cell viability, ACTH secretion, and Pomc expression. These effects were apparent at mitotane doses greater than those usually necessary for reducing cortisol secretion in Cushing's syndrome, but still in the therapeutic window for adrenocortical carcinoma treatment. In conclusion, our results demonstrate that mitotane affects cell viability and function of human and mouse ACTH-secreting pituitary adenoma cells. These data indicate that mitotane could have direct pituitary effects on corticotroph cells.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Via Savonarola 9, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gagliano T, Bellio M, Gentilin E, Molè D, Tagliati F, Schiavon M, Cavallesco NG, Andriolo LG, Ambrosio MR, Rea F, Degli Uberti E, Zatelli MC. mTOR, p70S6K, AKT, and ERK1/2 levels predict sensitivity to mTOR and PI3K/mTOR inhibitors in human bronchial carcinoids. Endocr Relat Cancer 2013; 20:463-75. [PMID: 23653462 DOI: 10.1530/erc-13-0042] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bronchial carcinoids (BCs) are rare neuroendocrine tumors that are still orphans of medical treatment. Human BC primary cultures may display resistance to everolimus, an inhibitor of the mammalian target of rapamycin (mTOR), in terms of cell viability reduction. Our aim was to assess whether the novel dual phosphatidylinositol 3-kinase (PI3K)/mTOR inhibitor NVP-BEZ235 is effective in everolimus-resistant human BC tissues and cell lines. In addition, we searched for possible markers of the efficacy of mTOR inhibitors that may help in identifying the patients who may benefit from treatment with mTOR inhibitors, sparing them from ineffective therapy. We found that NVP-BEZ235 is twice as potent as everolimus in reducing cell viability and activating apoptosis in human BC tissues that display sensitivity to mTOR inhibitors, but is not effective in everolimus-resistant BC tissues and cell lines that bypass cyclin D1 downregulation and escape G0/G1 blockade. Rebound AKT activation was not observed in response to treatment with either mTOR inhibitor in the 'resistant' BC cells. In addition to total mTOR levels, putative markers of the sensitivity of BCs to mTOR inhibitors are represented by AKT, p70S6K (RPS6KB2), and ERK1/2 (MAPK3/1) protein levels. Finally, we validated these markers in an independent BC group. These data indicate that the dual PI3K/mTOR inhibitor NVP-BEZ235 is more potent than everolimus in reducing the proliferation of human BC cells. 'Resistant' cells display lower levels of mTOR, p70S6K, AKT, and ERK1/2, indicating that these proteins may be useful as predictive markers of resistance to mTOR and PI3K/mTOR inhibitors in human BCs.
Collapse
Affiliation(s)
- Teresa Gagliano
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Via Savonarola 9, Ferrara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen X, Xue F, Xie T, Luo C. RETRACTED ARTICLE: Silencing of the Smad nuclear interacting protein 1 (SNIP1) by siRNA inhibits proliferation and induces apoptosis in pituitary adenoma cells. Tumour Biol 2013; 34:3071-6. [DOI: 10.1007/s13277-013-0873-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/16/2013] [Indexed: 11/29/2022] Open
|
25
|
Gentilin E, Tagliati F, Filieri C, Molè D, Minoia M, Rosaria Ambrosio M, Degli Uberti EC, Zatelli MC. miR-26a plays an important role in cell cycle regulation in ACTH-secreting pituitary adenomas by modulating protein kinase Cδ. Endocrinology 2013; 154:1690-700. [PMID: 23525216 PMCID: PMC3695590 DOI: 10.1210/en.2012-2070] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The functional aftermath of microRNA (miRNA) dysregulation in ACTH-secreting pituitary adenomas has not been demonstrated. miRNAs represent diagnostic and prognostic biomarkers as well as putative therapeutic targets; their investigation may shed light on the mechanisms that underpin pituitary adenoma development and progression. Drugs interacting with such pathways may help in achieving disease control also in the settings of ACTH-secreting pituitary adenomas. We investigated the expression of 10 miRNAs among those that were found as most dysregulated in human pituitary adenoma tissues in the settings of a murine ACTH-secreting pituitary adenoma cell line, AtT20/D16v-F2. The selected miRNAs to be submitted to further investigation in AtT20/D16v-F2 cells represent an expression panel including 5 up-regulated and 5 down-regulated miRNAs. Among these, we selected the most dysregulated mouse miRNA and searched for miRNA targets and their biological function. We found that AtT20/D16v-F2 cells have a specific miRNA expression profile and that miR-26a is the most dysregulated miRNA. The latter is overexpressed in human pituitary adenomas and can control viable cell number in the in vitro model without involving caspase 3/7-mediated apoptosis. We demonstrated that protein kinase Cδ (PRKCD) is a direct target of miR-26a and that miR26a inhibition delays the cell cycle in G1 phase. This effect involves down-regulation of cyclin E and cyclin A expression via PRKCD modulation. miR-26a and related pathways, such as PRKCD, play an important role in cell cycle control of ACTH pituitary cells, opening new therapeutic possibilities for the treatment of persistent/recurrent Cushing's disease.
Collapse
Affiliation(s)
- Erica Gentilin
- Section of Endocrinology, Department of Medical Sciences, University of Ferrara, Via Savonarola 9, 44100 Ferrara, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Short MK, Hallett JP, Tar K, Dange T, Schmidt M, Moir R, Willis IM, Jubinsky PT. The yeast magmas ortholog pam16 has an essential function in fermentative growth that involves sphingolipid metabolism. PLoS One 2012; 7:e39428. [PMID: 22808036 PMCID: PMC3393719 DOI: 10.1371/journal.pone.0039428] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Accepted: 05/22/2012] [Indexed: 01/06/2023] Open
Abstract
Magmas is a growth factor responsive gene encoding an essential mitochondrial protein in mammalian cells. Pam16, the Magmas ortholog in Saccharomyces cerevisiae, is a component of the presequence translocase-associated motor. A temperature-sensitive allele (pam16-I61N) was used to query an array of non-essential gene-deletion strains for synthetic genetic interactions. The pam16-I61N mutation at ambient temperature caused synthetic lethal or sick phenotypes with genes involved in lipid metabolism, perixosome synthesis, histone deacetylation and mitochondrial protein import. The gene deletion array was also screened for suppressors of the pam16-I61N growth defect to identify compensatory pathways. Five suppressor genes were identified (SUR4, ISC1, IPT1, SKN1, and FEN1) and all are involved in sphingolipid metabolism. pam16-I61N cells cultured in glucose at non-permissive temperatures resulted in rapid growth inhibition and G1 cell cycle arrest, but cell viability was maintained. Altered mitochondria morphology, reduced peroxisome induction in glycerol/ethanol and oleate, and changes in the levels of several sphingolipids including C18 alpha-hydroxy-phytoceramide, were also observed in the temperature sensitive strain. Deletion of SUR4, the strongest suppressor, reversed the temperature sensitive fermentative growth defect, the morphological changes and the elevated levels of C18 alpha-hydroxy phytoceramide in pam16-I61N. Deletion of the other four suppressor genes had similar effects on C18 alpha-hydroxy-phytoceramide levels and restored proliferation to the pam16-I61N strain. In addition, pam16-I61N inhibited respiratory growth, likely by reducing cardiolipin, which is essential for mitochondrial function. Our results suggest that the pleiotropic effects caused by impaired Pam16/Magmas function are mediated in part by changes in lipid metabolism.
Collapse
Affiliation(s)
- Mary K. Short
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Joshua P. Hallett
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Krisztina Tar
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Thomas Dange
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Marion Schmidt
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Robyn Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Paul T. Jubinsky
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Pediatrics, Section of Hematology/Oncology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
27
|
Roy S, Short MK, Stanley ER, Jubinsky PT. Essential role of Drosophila black-pearl is mediated by its effects on mitochondrial respiration. FASEB J 2012; 26:3822-33. [PMID: 22700875 DOI: 10.1096/fj.11-193540] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Black-pearl (Blp) is a highly conserved, essential inner-mitochondrial membrane protein. The yeast Blp homologue, Magmas/Pam16, is required for mitochondrial protein transport, growth, and survival. Our purpose was to determine the role of Drosophila Blp in mitochondrial function, cell survival, and proliferation. To this end, we performed mitotic recombination in Drosophila melanogaster, RNAi-mediated knockdown, MitoTracker staining, measurement of reactive oxygen species (ROS), flow cytometry, electron transport chain complex assays, and hemocyte isolation from Drosophila larvae. Proliferation-defective, Blp-deficient Drosophila Schneider cells exhibited mitochondrial membrane depolarization, a 60% decrease in ATP levels, increased amounts of ROS (3.5-fold), cell cycle arrest, and activation of autophagy that were associated with a selective 65% reduction of cytochrome c oxidase activity. N-acetyl cysteine (NAC) rescued Blp-RNAi-treated cells from cell cycle arrest, indicating that increased production of ROS is the primary cause of the proliferation and survival defects in Blp-depleted cells. blp hypomorph larvae had a 35% decreased number of plasmatocytes with a 45% reduced active mitochondrial staining and their viability was increased 2-fold by administration of NAC, which blocked melanotic lesions. Loss of Blp decreases cytochrome c oxidase activity and uncouples oxidative phosphorylation, causing ROS production, which selectively affects mitochondria-rich plasmatocyte survival and function, leading to melanotic lesions in Blp-deficient flies.
Collapse
Affiliation(s)
- Soumit Roy
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
28
|
Minoia M, Gentilin E, Molè D, Rossi M, Filieri C, Tagliati F, Baroni A, Ambrosio MR, degli Uberti E, Zatelli MC. Growth hormone receptor blockade inhibits growth hormone-induced chemoresistance by restoring cytotoxic-induced apoptosis in breast cancer cells independently of estrogen receptor expression. J Clin Endocrinol Metab 2012; 97:E907-16. [PMID: 22442272 DOI: 10.1210/jc.2011-3340] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT GH and IGF-I play a role in breast cancer (BC) development. We previously demonstrated that GH protects the estrogen receptor (ER) positive BC-derived MCF7 cell line toward the cytotoxic effects of doxorubicin (D), independently of IGF-I. This issue may be important in ER negative BC cells that are more aggressive and more likely to develop chemoresistance. AIM OF THE STUDY The aim of this study was to evaluate whether GH may impact chemoresistance phenotype of ER-negative BC-derived MDA-MB-231 cell line and investigate the possible mechanisms implicated in the protective action of GH toward the cytotoxic effects of D in both ER-positive and ER-negative BC-derived cell lines. RESULTS GH protects ER-negative MDA-MB-231 cells from the cytotoxic effects of D and GH receptor antagonist pegvisomant reduces GH-induced DNA synthesis also in these cells. In both MDA-MB-231 and MCF7 cells, GH does not revert D-induced G2/M accumulation but significantly reduces basal and D-induced apoptosis, an effect blocked by pegvisomant. Glutathione S-transferase activity is not implicated in the protective effects of GH, whereas D-induced apoptosis depends on c-Jun N terminal kinase (JNK) activation. GH reduces both basal and D-stimulated JNK transcriptional activity and phosphorylation. CONCLUSIONS In human BC cell lines, GH directly promotes resistance to apoptosis induced by chemotherapeutic drugs independently of ER expression by modulating JNK, further broadening the concept that GH excess may hamper cytotoxic BC treatment. These findings support the hypothesis that blocking GH receptor may be viewed as a potential new therapeutic approach to overcome chemoresistance, especially in ER-negative BC.
Collapse
Affiliation(s)
- Mariella Minoia
- Section of Endocrinology, Department of Biomedical Sciences and Advanced Therapies, University of Ferrara, Via Savonarola 9, 44121 Ferrara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
[Neuroendocrinology in 2011]. ENDOCRINOLOGIA Y NUTRICION : ORGANO DE LA SOCIEDAD ESPANOLA DE ENDOCRINOLOGIA Y NUTRICION 2012; 59:311-25. [PMID: 22425316 DOI: 10.1016/j.endonu.2012.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 01/04/2023]
|
30
|
Jubinsky PT, Short MK, Ghanem M, Das BC. Design, synthesis, and biological activity of novel Magmas inhibitors. Bioorg Med Chem Lett 2011; 21:3479-82. [PMID: 21514823 DOI: 10.1016/j.bmcl.2011.03.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 03/10/2011] [Accepted: 03/14/2011] [Indexed: 11/18/2022]
Abstract
Magmas (mitochondria associated, granulocyte-macrophage colony stimulating factor signaling molecule), is a highly conserved and essential gene, expressed in all cell types. We designed and synthesized several small molecule Magmas inhibitors (SMMI) and assayed their effects on proliferation in yeast. We found that the most active compound 9 inhibited growth at the 4 μM scale. This compound was shown by fluorometric titration to bind to Magmas with a K(d)=33 μM. Target specificity of the lead compound was established by demonstrating direct binding of the compound to Magmas and by genetic studies. Molecular modeling suggested that the inhibitor bound at the predicted site in Magmas.
Collapse
Affiliation(s)
- Paul T Jubinsky
- Pediatric Hematology/Oncology, Yale University Medical School, New Haven, CT 06510, United States.
| | | | | | | |
Collapse
|