1
|
Barzilai DA. Mikhail 'Misha' Blagosklonny's enduring legacy in geroscience: the hyperfunction theory and the therapeutic potential of rapamycin. Aging (Albany NY) 2025; null:206189. [PMID: 39808121 DOI: 10.18632/aging.206189] [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: 10/31/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
The untimely passing of Dr. Mikhail "Misha" Blagosklonny has left a lasting void in geroscience and oncology. This review examines his profound contributions, focusing on his pioneering the Hyperfunction Theory and his advocacy for rapamycin, an mTOR inhibitor, as a therapeutic agent for lifespan extension. Contrary to traditional damage-centric models, the Hyperfunction Theory rejects damage accumulation as the primary driver of aging. Instead, it redefines aging as a quasi-programmed process driven by the persistent, excessive activity of growth-promoting pathways beyond their developmental roles, leading to age-related pathologies. We explore how Blagosklonny's insights predict rapamycin's ability to decelerate aging by modulating excessive mTOR signaling, supported by empirical evidence across multiple physiological systems, including immune, cardiovascular, cognitive, and oncologic health. His forward-thinking approach, advocating for the cautious clinical use of rapamycin and suggesting personalized, preventive, and combination therapy strategies, has catalyzed interest in translational geroscience. This review synthesizes Blagosklonny's legacy, presenting rapamycin as a foundational pharmacological intervention with potential in managing age-related decline and extending healthspan, and underlines his impact in shifting aging research from theoretical frameworks to actionable interventions. Blagosklonny's work remains an enduring inspiration, paving the way toward treating aging as a modifiable condition.
Collapse
Affiliation(s)
- David A Barzilai
- Geneva College of Longevity Science, Geneva 1204, Switzerland
- Healthspan Coaching LLC, Barzilai Longevity Consulting, Boston, MA 02111, USA
| |
Collapse
|
2
|
Libalova H, Zavodna T, Margaryan H, Elzeinova F, Milcova A, Vrbova K, Barosova H, Cervena T, Topinka J, Rössner P. Differential DNA damage response and cell fate in human lung cells after exposure to genotoxic compounds. Toxicol In Vitro 2024; 94:105710. [PMID: 37838151 DOI: 10.1016/j.tiv.2023.105710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
DNA damage can impair normal cellular functions and result in various pathophysiological processes including cardiovascular diseases and cancer. We compared the genotoxic potential of diverse DNA damaging agents, and focused on their effects on the DNA damage response (DDR) and cell fate in human lung cells BEAS-2B. Polycyclic aromatic hydrocarbons [PAHs; benzo[a]pyrene (B[a]P), 1-nitropyrene (1-NP)] induced DNA strand breaks and oxidative damage to DNA; anticancer drugs doxorubicin (DOX) and 5-bromo-2'-deoxyuridine (BrdU) were less effective. DOX triggered the most robust p53 signaling indicating activation of DDR, followed by cell cycle arrest in the G2/M phase, induction of apoptosis and senescence, possibly due to the severe and irreparable DNA lesions. BrdU not only activated p53, but also increased the percentage of G1-phased cells and caused a massive accumulation of senescent cells. In contrast, regardless the activation of p53, both PAHs did not substantially affect the cell cycle distribution or senescence. Finally, a small fraction of cells accumulated only in the G2/M phase and exhibited increased cell death after the prolonged incubation with B[a]P. Overall, we characterized differential responses to diverse DNA damaging agents resulting in specific cell fate and highlighted the key role of DNA lesion type and the p53 signaling persistence.
Collapse
Affiliation(s)
- H Libalova
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - T Zavodna
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - H Margaryan
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - F Elzeinova
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - A Milcova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - K Vrbova
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - H Barosova
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - T Cervena
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic; Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - J Topinka
- Department of Genetic Toxicology and Epigenetics, Institute of Experimental Medicine of the CAS, Prague, Czech Republic
| | - P Rössner
- Department of Nanotoxicology and Molecular Epidemiology, Institute of Experimental Medicine of the CAS, Prague, Czech Republic.
| |
Collapse
|
3
|
Blagosklonny MV. As expected, based on rapamycin-like p53-mediated gerosuppression, mTOR inhibition acts as a checkpoint in p53-mediated tumor suppression. Oncoscience 2022; 9:38-41. [PMID: 36052376 PMCID: PMC9426927 DOI: 10.18632/oncoscience.561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
|
4
|
Yang S, Han G, Chen Q, Yu L, Wang P, Zhang Q, Dong J, Zhang W, Huang J. Au-Pt Nanoparticle Formulation as a Radiosensitizer for Radiotherapy with Dual Effects. Int J Nanomedicine 2021; 16:239-248. [PMID: 33469284 PMCID: PMC7811476 DOI: 10.2147/ijn.s287523] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Background Radiotherapy occupies an essential position as one of the most significant approaches for the clinical treatment of cancer. However, we cannot overcome the shortcoming of X-rays which is the high value of the oxygen enhancement ratio (OER). Radiosensitizers with the ability to enhance the radiosensitivity of tumor cells provide an alternative to changing X-rays to protons and heavy ion radiotherapy. Materials and Methods We prepared the Au-Pt nanoparticles (Au-Pt NPs) using a one-step method. The characteristics of the Au-Pt NPs were determined using TEM, HAADF-STEM, elemental mapping images, and DLS. The enhanced radiotherapy was demonstrated in vitro using MTT assays, colony formation assays, fluorescence imaging, and flow cytometric analyses of the apoptosis. The biodistribution of the Au-Pt NPs was analyzed using ICP-OES, and thermal images. The enhanced radiotherapy was demonstrated in vitro using immunofluorescence images, tumor volume and weigh, and hematoxylin & eosin (H&E) staining. Results Polyethylene glycol (PEG) functionalized nanoparticles composed of the metallic elements Au and Pt were designed to increase synergistic radiosensitivity. The mechanism demonstrated that heavy metal NPs possess a high X-ray photon capture cross-section and Compton scattering effect which increased DNA damage. Furthermore, the Au-Pt NPs exhibited enzyme-mimicking activities by catalyzing the decomposition of endogenous H2O2 to O2 in the solid tumor microenvironment (TME). Conclusion Our work provides a systematically administered radiosensitizer that can selectively reside in a tumor via the EPR effect and enhances the efficiency of treating cancer with radiotherapy.
Collapse
Affiliation(s)
- Song Yang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Gaohua Han
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Quan Chen
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Thoracic Surgery, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Lei Yu
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Peng Wang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qi Zhang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jiang Dong
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Wei Zhang
- Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China.,Department of Infectious Disease, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China
| | - Junxing Huang
- Department of Oncology, Taizhou People's Hospital, Taizhou, Jiangsu, People's Republic of China.,Medical School of Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
5
|
Lee JY, Kennedy BK, Liao CY. Mechanistic target of rapamycin signaling in mouse models of accelerated aging. J Gerontol A Biol Sci Med Sci 2020; 75:64-72. [PMID: 30900725 DOI: 10.1093/gerona/glz059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/23/2019] [Indexed: 01/06/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) is an essential nutrient-sensing kinase that integrates and regulates a number of fundamental cellular processes required for cell growth, cell motility, translation, metabolism, and autophagy. mTOR signaling has been implicated in the progression of many human diseases, and its dysregulation has been reported in several pathological processes, especially in age-related human diseases and mouse models of accelerated aging. In addition, many studies have demonstrated that the regulation of mTOR activity has a beneficial effect on longevity in several mouse models of aging. However, not all mouse models of accelerated aging show positive effects on aging-associated phenotypes in response to targeting mTOR signaling. Here, we review the effects of interventions that modulate mTOR signaling on aging-related phenotypes in different mouse models of accelerated aging and discuss their implications with respect to aging and aging-related disorders.
Collapse
Affiliation(s)
- Jin Young Lee
- Buck Institute for Research on Aging, Novato, California
| | - Brian K Kennedy
- Buck Institute for Research on Aging, Novato, California
- Department of Biochemistry and Physiology, National University of Singapore, Singapore
- Centre for Healthy Ageing, National University Health System, Singapore
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
| | - Chen-Yu Liao
- Buck Institute for Research on Aging, Novato, California
| |
Collapse
|
6
|
Bezzerri V, Piacenza F, Caporelli N, Malavolta M, Provinciali M, Cipolli M. Is cellular senescence involved in cystic fibrosis? Respir Res 2019; 20:32. [PMID: 30764828 PMCID: PMC6376730 DOI: 10.1186/s12931-019-0993-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary disease is the main cause of the morbidity and mortality of patients affected by cystic fibrosis (CF). The lung pathology is dominated by excessive recruitment of neutrophils followed by an exaggerated inflammatory process that has also been reported to occur in the absence of apparent pathogenic infections. Airway surface dehydration and mucus accumulation are the driving forces of this process. The continuous release of reactive oxygen species and proteases by neutrophils contributes to tissue damage, which eventually leads to respiratory insufficiency. CF has been considered a paediatric problem for several decades. Nevertheless, during the last 40 years, therapeutic options for CF have been greatly improved, turning CF into a chronic disease and extending the life expectancy of patients. Unfortunately, chronic inflammatory processes, which are characterized by a substantial release of cytokines and chemokines, along with ROS and proteases, can accelerate cellular senescence, leading to further complications in adulthood. The alterations and mechanisms downstream of CFTR functional defects that can stimulate cellular senescence remain unclear. However, while there are correlative data suggesting that cellular senescence may be implicated in CF, a causal or consequential relationship between cellular senescence and CF is still far from being established. Senescence can be both beneficial and detrimental. Senescence may suppress bacterial infections and cooperate with tissue repair. Additionally, it may act as an effective anticancer mechanism. However, it may also promote a pro-inflammatory environment, thereby damaging tissues and leading to chronic age-related diseases. In this review, we present the most current knowledge on cellular senescence and contextualize its possible involvement in CF.
Collapse
Affiliation(s)
- Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Nicole Caporelli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Marco Cipolli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy.
| |
Collapse
|
7
|
p53 Functions in Adipose Tissue Metabolism and Homeostasis. Int J Mol Sci 2018; 19:ijms19092622. [PMID: 30181511 PMCID: PMC6165290 DOI: 10.3390/ijms19092622] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/24/2018] [Accepted: 08/30/2018] [Indexed: 12/25/2022] Open
Abstract
As a tumor suppressor and the most frequently mutated gene in cancer, p53 is among the best-described molecules in medical research. As cancer is in most cases an age-related disease, it seems paradoxical that p53 is so strongly conserved from early multicellular organisms to humans. A function not directly related to tumor suppression, such as the regulation of metabolism in nontransformed cells, could explain this selective pressure. While this role of p53 in cellular metabolism is gradually emerging, it is imperative to dissect the tissue- and cell-specific actions of p53 and its downstream signaling pathways. In this review, we focus on studies reporting p53’s impact on adipocyte development, function, and maintenance, as well as the causes and consequences of altered p53 levels in white and brown adipose tissue (AT) with respect to systemic energy homeostasis. While whole body p53 knockout mice gain less weight and fat mass under a high-fat diet owing to increased energy expenditure, modifying p53 expression specifically in adipocytes yields more refined insights: (1) p53 is a negative regulator of in vitro adipogenesis; (2) p53 levels in white AT are increased in diet-induced and genetic obesity mouse models and in obese humans; (3) functionally, elevated p53 in white AT increases senescence and chronic inflammation, aggravating systemic insulin resistance; (4) p53 is not required for normal development of brown AT; and (5) when p53 is activated in brown AT in mice fed a high-fat diet, it increases brown AT temperature and brown AT marker gene expression, thereby contributing to reduced fat mass accumulation. In addition, p53 is increasingly being recognized as crucial player in nutrient sensing pathways. Hence, despite existence of contradictory findings and a varying density of evidence, several functions of p53 in adipocytes and ATs have been emerging, positioning p53 as an essential regulatory hub in ATs. Future studies need to make use of more sophisticated in vivo model systems and should identify an AT-specific set of p53 target genes and downstream pathways upon different (nutrient) challenges to identify novel therapeutic targets to curb metabolic diseases.
Collapse
|
8
|
Vazquez-Martin A, Anatskaya OV, Giuliani A, Erenpreisa J, Huang S, Salmina K, Inashkina I, Huna A, Nikolsky NN, Vinogradov AE. Somatic polyploidy is associated with the upregulation of c-MYC interacting genes and EMT-like signature. Oncotarget 2018; 7:75235-75260. [PMID: 27655693 PMCID: PMC5342737 DOI: 10.18632/oncotarget.12118] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 09/05/2016] [Indexed: 12/30/2022] Open
Abstract
The dependence of cancer on overexpressed c-MYC and its predisposition for polyploidy represents a double puzzle. We address this conundrum by cross-species transcription analysis of c-MYC interacting genes in polyploid vs. diploid tissues and cells, including human vs. mouse heart, mouse vs. human liver and purified 4n vs. 2n mouse decidua cells. Gene-by-gene transcriptome comparison and principal component analysis indicated that c-MYC interactants are significantly overrepresented among ploidy-associated genes. Protein interaction networks and gene module analysis revealed that the most upregulated genes relate to growth, stress response, proliferation, stemness and unicellularity, as well as to the pathways of cancer supported by MAPK and RAS coordinated pathways. A surprising feature was the up-regulation of epithelial-mesenchymal transition (EMT) modules embodied by the N-cadherin pathway and EMT regulators from SNAIL and TWIST families. Metabolic pathway analysis also revealed the EMT-linked features, such as global proteome remodeling, oxidative stress, DNA repair and Warburg-like energy metabolism. Genes associated with apoptosis, immunity, energy demand and tumour suppression were mostly down-regulated. Noteworthy, despite the association between polyploidy and ample features of cancer, polyploidy does not trigger it. Possibly it occurs because normal polyploidy does not go that far in embryonalisation and linked genome destabilisation. In general, the analysis of polyploid transcriptome explained the evolutionary relation of c-MYC and polyploidy to cancer.
Collapse
Affiliation(s)
| | - Olga V Anatskaya
- Institute of Cytology, St-Petersburg, Russian Federation, Russia
| | | | | | - Sui Huang
- Systems Biology Institute, Seattle, USA
| | | | - Inna Inashkina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Anda Huna
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | |
Collapse
|
9
|
Abstract
Senescence contributes to the local and systemic aging of tissues and has been associated with age-related diseases. Recently, roles for this process during pregnancy have come to light, the dysregulation of which has been associated with adverse pregnancy outcomes such as preterm birth. Here, we summarize recent advances that support a role for senescence in birth timing and propose new aspects of study in this emerging field.
Collapse
Affiliation(s)
- Jeeyeon M Cha
- a Division of Diabetes, Endocrinology and Metabolism , Vanderbilt University Medical Center , Nashville , TN , USA.,b Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA
| | - David M Aronoff
- b Department of Medicine , Vanderbilt University Medical Center , Nashville , TN , USA.,c Division of Infectious Diseases , Vanderbilt University Medical Center , Nashville , TN , USA.,d Department of Pathology, Microbiology and Immunology , Vanderbilt University Medical Center , Nashville , TN , USA
| |
Collapse
|
10
|
Zining J, Lu X, Caiyun H, Yuan Y. Genetic polymorphisms of mTOR and cancer risk: a systematic review and updated meta-analysis. Oncotarget 2016; 7:57464-57480. [PMID: 27462867 PMCID: PMC5302868 DOI: 10.18632/oncotarget.10805] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/29/2016] [Indexed: 12/16/2022] Open
Abstract
mTOR regulates several cellular processes that are critical for tumorigenesis. However, previous studies on the association of mTOR polymorphisms with predisposition to different cancer types are somewhat contradictory. Therefore, we performed a systematic review and updated meta-analysis of the available evidence regarding the relationship between mTOR single nucleotide polymorphisms (SNPs) and cancer risk. Up to November 2015, 23 original publications were identified covering 20 mTOR SNPs, of which seven SNPs (rs2536, rs2295080, rs1883965, rs1034528, rs17036508, rs3806317 and rs1064261) were included in the final meta-analysis. We estimated the summary odds ratios (ORs) and corresponding 95% confidence intervals (CIs) for mTOR polymorphisms and cancer risk, and used the model-free approach to investigate the biological effect of each polymorphism. Our meta-analysis found that rs1883965, rs1034528, and rs17036508 were correlated with increased cancer risk in the complete over-dominant model (rs1883965 GA versus GG/AA: fixed-effects OR=1.15, 95% CI 1.02-1.29; rs1034528 GC versus GG/CC: fixed-effects OR=1.30, 95% CI 1.13-1.48; rs17036508 TC versus CC/TT: fixed-effects OR=1.23, 95% CI 1.06-1.43). Stratifying analyses by cancer type, we found that the rs2295080 G allele was associated with a significantly higher risk of acute leukemia in the recessive model (GG versus GT/TT: fixed-effects OR=2.08, 95% CI 1.34-3.22) and a lower risk of genitourinary cancers in the dominant model (TG/GG versus TT: fixed-effects OR=0.77, 95% CI 0.68-0.86). Interestingly, further expression analysis showed that homozygous variant genotype carriers of rs1883965, rs1034528 and rs17036508 had lower mTOR transcript levels, based on HapMap data.
Collapse
Affiliation(s)
- Jin Zining
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention of Liaoning Provincial Education Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xu Lu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention of Liaoning Provincial Education Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - He Caiyun
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, Key Laboratory of Cancer Etiology and Prevention of Liaoning Provincial Education Department, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Xiong M, Ferder IC, Ohguchi Y, Wang N. Quantitative analysis of male germline stem cell differentiation reveals a role for the p53-mTORC1 pathway in spermatogonial maintenance. Cell Cycle 2016; 14:2905-13. [PMID: 26177380 DOI: 10.1080/15384101.2015.1069928] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
p53 protects cells from DNA damage by inducing cell-cycle arrest upon encountering genomic stress. Among other pathways, p53 elicits such an effect by inhibiting mammalian target of rapamycin complex 1 (mTORC1), the master regulator of cell proliferation and growth. Although recent studies have indicated roles for both p53 and mTORC1 in stem cell maintenance, it remains unclear whether the p53-mTORC1 pathway is conserved to mediate this process under normal physiological conditions. Spermatogenesis is a classic stem cell-dependent process in which undifferentiated spermatogonia undergo self-renewal and differentiation to maintain the lifelong production of spermatozoa. To better understand this process, we have developed a novel flow cytometry (FACS)-based approach that isolates spermatogonia at consecutive differentiation stages. By using this as a tool, we show that genetic loss of p53 augments mTORC1 activity during early spermatogonial differentiation. Functionally, loss of p53 drives spermatogonia out of the undifferentiated state and causes a consistent expansion of early differentiating spermatogonia until the stage of preleptotene (premeiotic) spermatocyte. The frequency of early meiotic spermatocytes is, however, dramatically decreased. Thus, these data suggest that p53-mTORC1 pathway plays a critical role in maintaining the homeostasis of early spermatogonial differentiation. Moreover, our FACS approach could be a valuable tool in understanding spermatogonial differentiation.
Collapse
Affiliation(s)
- Mulin Xiong
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Ianina C Ferder
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Yasuyo Ohguchi
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| | - Ning Wang
- a Vincent Center for Reproductive Biology; Vincent Department of Obstetrics and Gynecology; Massachusetts General Hospital; Harvard Medical School ; Boston , MA USA
| |
Collapse
|
12
|
Leontieva OV, Demidenko ZN, Blagosklonny MV. Dual mTORC1/C2 inhibitors suppress cellular geroconversion (a senescence program). Oncotarget 2016; 6:23238-48. [PMID: 26177051 PMCID: PMC4695114 DOI: 10.18632/oncotarget.4836] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 07/24/2015] [Indexed: 11/30/2022] Open
Abstract
In proliferating cells, mTOR is active and promotes cell growth. When the cell cycle is arrested, then mTOR converts reversible arrest to senescence (geroconversion). Rapamycin and other rapalogs suppress geroconversion, maintaining quiescence instead. Here we showed that ATP-competitive kinase inhibitors (Torin1 and PP242), which inhibit both mTORC1 and TORC2, also suppressed geroconversion. Despite inhibition of proliferation (in proliferating cells), mTOR inhibitors preserved re-proliferative potential (RP) in arrested cells. In p21-arrested cells, Torin 1 and PP242 detectably suppressed geroconversion at concentrations as low as 1-3 nM and 10-30 nM, reaching maximal gerosuppression at 30 nM and 300 nM, respectively. Near-maximal gerosuppression coincided with inhibition of p-S6K(T389) and p-S6(S235/236). Dual mTOR inhibitors prevented senescent morphology and hypertrophy. Our study warrants investigation into whether low doses of dual mTOR inhibitors will prolong animal life span and delay age-related diseases. A new class of potential anti-aging drugs can be envisioned.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Zoya N Demidenko
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
13
|
Rejuvenating immunity: "anti-aging drug today" eight years later. Oncotarget 2016; 6:19405-12. [PMID: 25844603 PMCID: PMC4637294 DOI: 10.18632/oncotarget.3740] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/28/2015] [Indexed: 01/02/2023] Open
Abstract
The 2014 year ended with celebration: Everolimus, a rapamycin analog, was shown to improve immunity in old humans, heralding ‘a turning point’ in research and new era in human quest for immortality. Yet, this turning point was predicted a decade ago. But what will cause human death, when aging will be abolished?
Collapse
|
14
|
Christy B, Demaria M, Campisi J, Huang J, Jones D, Dodds SG, Williams C, Hubbard G, Livi CB, Gao X, Weintraub S, Curiel T, Sharp ZD, Hasty P. p53 and rapamycin are additive. Oncotarget 2016; 6:15802-13. [PMID: 26158292 PMCID: PMC4599238 DOI: 10.18632/oncotarget.4602] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/14/2015] [Indexed: 12/13/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a kinase found in a complex (mTORC1) that enables macromolecular synthesis and cell growth and is implicated in cancer etiology. The rapamycin-FK506 binding protein 12 (FKBP12) complex allosterically inhibits mTORC1. In response to stress, p53 inhibits mTORC1 through a separate pathway involving cell signaling and amino acid sensing. Thus, these different mechanisms could be additive. Here we show that p53 improved the ability of rapamycin to: 1) extend mouse life span, 2) suppress ionizing radiation (IR)-induced senescence-associated secretory phenotype (SASP) and 3) increase the levels of amino acids and citric acid in mouse embryonic stem (ES) cells. This additive effect could have implications for cancer treatment since rapamycin and p53 are anti-oncogenic.
Collapse
Affiliation(s)
- Barbara Christy
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Marco Demaria
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Jing Huang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Diane Jones
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sherry G Dodds
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Charnae Williams
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Gene Hubbard
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Carolina B Livi
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Current address: Agilent Technologies, Inc., Santa Clara, CA, USA
| | - Xiaoli Gao
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Susan Weintraub
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tyler Curiel
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Z Dave Sharp
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Paul Hasty
- Departments of Molecular Medicine and Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
15
|
Leontieva OV, Blagosklonny MV. Tumor promoter-induced cellular senescence: cell cycle arrest followed by geroconversion. Oncotarget 2015; 5:12715-27. [PMID: 25587030 PMCID: PMC4350340 DOI: 10.18632/oncotarget.3011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 12/26/2014] [Indexed: 02/07/2023] Open
Abstract
Phorbol ester (PMA or TPA), a tumor promoter, can cause either proliferation or cell cycle arrest, depending on cellular context. For example, in SKBr3 breast cancer cells, PMA hyper-activates the MEK/MAPK pathway, thus inducing p21 and cell cycle arrest. Here we showed that PMA-induced arrest was followed by conversion to cellular senescence (geroconversion). Geroconversion was associated with active mTOR and S6 kinase (S6K). Rapamycin suppressed geroconversion, maintaining quiescence instead. In this model, PMA induced arrest (step one of a senescence program), whereas constitutively active mTOR drove geroconversion (step two). Without affecting Akt phosphorylation, PMA increased phosphorylation of S6K (T389) and S6 (S240/244), and that was completely prevented by rapamycin. Yet, T421/S424 and S235/236 (p-S6K and p-S6, respectively) phosphorylation became rapamycin-insensitive in the presence of PMA. Either MEK or mTOR was sufficient to phosphorylate these PMA-induced rapamycin-resistant sites because co-treatment with U0126 and rapamycin was required to abrogate them. We next tested whether activation of rapamycin-insensitive pathways would shift quiescence towards senescence. In HT-p21 cells, cell cycle arrest was caused by IPTG-inducible p21 and was spontaneously followed by mTOR-dependent geroconversion. Rapamycin suppressed geroconversion, whereas PMA partially counteracted the effect of rapamycin, revealing the involvement of rapamycin-insensitive gerogenic pathways. In normal RPE cells arrested by serum withdrawal, the mTOR/pS6 pathway was inhibited and cells remained quiescent. PMA transiently activated mTOR, enabling partial geroconversion. We conclude that PMA can initiate a senescent program by either inducing arrest or fostering geroconversion or both. Rapamycin can decrease gero-conversion by PMA, without preventing PMA-induced arrest. The tumor promoter PMA is a gero-promoter, which may be useful to study aging in mammals.
Collapse
Affiliation(s)
- Olga V Leontieva
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | | |
Collapse
|
16
|
Landuzzi L, Ianzano ML, Nicoletti G, Palladini A, Grosso V, Ranieri D, Dall'Ora M, Raschi E, Laranga R, Gambarotti M, Picci P, De Giovanni C, Nanni P, Lollini PL. Genetic prevention of lymphoma in p53 knockout mice allows the early development of p53-related sarcomas. Oncotarget 2015; 5:11924-38. [PMID: 25426555 PMCID: PMC4322986 DOI: 10.18632/oncotarget.2650] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/27/2014] [Indexed: 11/25/2022] Open
Abstract
Homozygous knockout of p53 in mice leads to early mortality from lymphoma, with almost complete penetrance, thus hampering studies of other tumor histotypes related to p53 alterations. To avoid lymphoma development, we crossed p53 knockout mice (BALB-p53 mice) with alymphocytic BALB/c Rag2−/−;Il2rg−/− (RGKO) mice. We compared the tumor spectrum of homozygous (BALB-p53−/−) and heterozygous (BALB-p53+/−) mice with alymphocytic mice (RGKO-p53−/− and RGKO-p53+/−). Lymphoma incidence in BALB-p53−/− mice exceeded 80%, whereas in RGKO-p53−/− it was strongly reduced. The prevalent tumor of RGKO-p53−/− mice was hemangiosarcoma (incidence over 65% in both sexes, mean latency 18 weeks), other tumors included soft tissue sarcomas (incidence ~10%), lung and mammary carcinomas. Tumor spectrum changes occurred also in p53 heterozygotes, in which lymphomas are relatively rare (~20%). RGKO-p53+/− had an increased incidence of hemangiosarcomas, reaching ~30%, and females had an increased incidence of osteosarcomas, reaching ~20%. Osteosarcomas shared with the corresponding human tumors the involvement of limbs and a high metastatic ability, mainly to the lungs. Specific alterations in the expression of p53-related genes (p16Ink4a, p19Arf, p15Ink4b, p21Cip1) were observed. Genetic prevention of lymphoma in p53 knockout mice led to new models of sarcoma development, available for studies on hemangiosarcoma and osteosarcoma onset and metastatization.
Collapse
Affiliation(s)
- Lorena Landuzzi
- Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Bologna, Italy. PROMETEO Laboratory, STB, RIT Department, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Marianna L Ianzano
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Giordano Nicoletti
- Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Bologna, Italy. PROMETEO Laboratory, STB, RIT Department, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Valentina Grosso
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Dario Ranieri
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Massimiliano Dall'Ora
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Elena Raschi
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Roberta Laranga
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Marco Gambarotti
- Anatomy and Pathological Histology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Piero Picci
- Laboratory of Experimental Oncology, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Carla De Giovanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna Italy
| |
Collapse
|
17
|
Abstract
Cellular senescence happens in 2 steps: cell cycle arrest followed, or sometimes preceded, by gerogenic conversion (geroconversion). Geroconvesrion is a form of growth, a futile growth during cell cycle arrest. It converts reversible arrest to irreversible senescence. Geroconversion is driven by growth-promoting, mitogen-/nutrient-sensing pathways such as mTOR. Geroconversion leads to hyper-secretory, hypertrophic and pro-inflammatory cellular phenotypes, hyperfunctions and malfunctions. On organismal level, geroconversion leads to age-related diseases and death. Rapamycin, a gerosuppressant, extends life span in diverse species from yeast to mammals. Stress-and oncogene-induced accelerated senescence, replicative senescence in vitro and life-long cellular aging in vivo all can be described by 2-step model.
Collapse
|
18
|
Ip CKM, Yung S, Chan TM, Tsao SW, Wong AST. p70 S6 kinase drives ovarian cancer metastasis through multicellular spheroid-peritoneum interaction and P-cadherin/b1 integrin signaling activation. Oncotarget 2015; 5:9133-49. [PMID: 25193855 PMCID: PMC4253424 DOI: 10.18632/oncotarget.2362] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Peritoneal dissemination as a manifestation of ovarian cancer is an adverse prognostic factor associated with poor clinical outcome, and is thus a potentially promising target for improved treatment. Sphere forming cells (multicellular spheroids) present in malignant ascites of patients with ovarian cancer represent a major impediment to effective treatment. p70 S6 kinase (p70S6K), which is a downstream effector of mammalian target of rapamycin, is frequently hyperactivated in human ovarian cancer. Here, we identified p70S6K as an important regulator for the seeding and successful colonization of ovarian cancer spheroids on the peritoneum. Furthermore, we provided evidence for the existence of a novel crosstalk between P-cadherin and β1 integrin, which was crucial for the high degree of specificity in cell adhesion. In particular, we demonstrated that the upregulation of mature β1 integrin occurred as a consequence of P-cadherin expression through the induction of the Golgi glycosyltransferase, ST6Gal-I, which mediated β1 integrin hypersialylation. Loss of p70S6K or targeting the P-cadherin/β1-integrin interplay could significantly attenuate the metastatic spread onto the peritoneum in vivo. These findings establish a new role for p70S6K in tumor spheroid-mesothelium communication in ovarian cancer and provide a preclinical rationale for targeting p70S6K as a new avenue for microenvironment-based therapeutic strategy.
Collapse
Affiliation(s)
- Carman Ka Man Ip
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong
| | - Susan Yung
- Department of Medicine, University of Hong Kong, Sassoon Road, Hong Kong
| | - Tak-Mao Chan
- Department of Medicine, University of Hong Kong, Sassoon Road, Hong Kong
| | - Sai-Wah Tsao
- Department of Anatomy, University of Hong Kong, Sassoon Road, Hong Kong
| | - Alice Sze Tsai Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong
| |
Collapse
|
19
|
Vici P, Sperati F, Maugeri-Saccà M, Melucci E, Di Benedetto A, Di Lauro L, Pizzuti L, Sergi D, Terrenato I, Esposito L, Iannuzzi CA, Pasquale R, Botti C, Fuhrman B, Giordano A, Mottolese M, Barba M. p53 status as effect modifier of the association between pre-treatment fasting glucose and breast cancer outcomes in non diabetic, HER2 positive patients treated with trastuzumab. Oncotarget 2015; 5:10382-92. [PMID: 25071015 PMCID: PMC4279380 DOI: 10.18632/oncotarget.2060] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/04/2014] [Indexed: 12/13/2022] Open
Abstract
Mounting evidence supports the role of p53 in metabolic processes involved in breast carcinogenesis. We investigated whether p53 status affects the association of pre-treatment fasting glucose with treatment outcomes in 106 non diabetic, HER2 positive breast cancer patients treated with trastuzumab. p53 status was validated against gene sequencing of selected codons in 49 patients. The Kaplan-Meier method and log rank test were used to compare survival by categories of fasting glucose in the overall population and separate settings. Cox models included age and body mass index. Direct sequencing confirmed the lack of mutations in 73.7% of p53 negative patients and their presence in 53.3% of p53 positive cases. At 66 months, 88.3% of patients with glucose ≤ 89.0 mg/dl (median value) did not experiment disease progression compared with 70.0% in the highest category (p=0.034), with glucose being an independent predictor (p=0.046). Stratified analysis confirmed this association in p53 negative patients only (p=0.01). In the early setting, data suggested longer disease free survival in p53 negative patients in the lowest glucose category (p=0.053). In our study, p53 status acted as effect modifier of the investigated association. This may help differentiate target sub-groups and affect outcomes interpretation in similarly characterized patients.
Collapse
Affiliation(s)
- Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology B-Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Di Benedetto
- Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| | - Luca Esposito
- Center for Oncologic Research of Mercogliano (CROM), Avellino, Italy
| | | | - Raffaella Pasquale
- Oncology Research Centre of Mercogliano (CROM), G. Pascale Foundation National Cancer Institute, Naples, Italy
| | - Claudio Botti
- Department of Surgery, Regina Elena National Cancer Institute, Rome, Italy
| | - Barbara Fuhrman
- Department of Epidemiology, University of Arkansas for Medical Sciences, Arkansas, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology Temple University, Philadelphia, USA
| | - Marcella Mottolese
- Department of Pathology, Regina Elena National Cancer Institute, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology B-Scientific Direction, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
20
|
Liu WL, Gao M, Tzen KY, Tsai CL, Hsu FM, Cheng AL, Cheng JCH. Targeting Phosphatidylinositide3-Kinase/Akt pathway by BKM120 for radiosensitization in hepatocellular carcinoma. Oncotarget 2015; 5:3662-72. [PMID: 25004403 PMCID: PMC4116511 DOI: 10.18632/oncotarget.1978] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tumor control of hepatocellular carcinoma by radiotherapy remains unsatisfactory. The phosphatidylinositol 3-kinase (PI3K)/Akt pathway plays a critical role in inhibiting cancer cell death. Elevated PI3K/Akt activity is associated with increased cellular resistance to irradiation. Our aim was to determine whether the inhibition of PI3K/Akt activity by a PI3K inhibitor, BKM120, contributes to the increased sensitivity of liver cancer cells to irradiation. The hepatocellular carcinoma cell lines (Huh7 and BNL) were used to evaluate the in vitro synergism between BKM120 and irradiation. Balb/c mice bearing ectopic BNL xenografts were treated with BKM120 and/or radiotherapy to assess the in vivo response. BKM120 increased cell killing by radiation, increased the expression of apoptotic markers, and suppressed the repair of radiation-induced DNA double-strand breaks. BKM120 pretreatment inhibited radiation-induced Akt phosphorylation and enhanced the tumor-suppressive effect and radiation-induced tumor cell apoptosis in ectopic xenografts. Inhibition of mTOR phosphorylation by rapamycin enhanced the radiosensitivity of BKM120-treated hepatocellular carcinoma cells. The synergism between BKM120 and irradiation likely inhibits the activation of Akt by radiation, leading to increased cell apoptosis and suppression of DNA-double-strand breaks repair in hepatocellular carcinoma cells. These data suggest that the BKM120/radiation combination may be a strategy worthy of clinical trials.
Collapse
Affiliation(s)
- Wei-Lin Liu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | - Jason Chia-Hsien Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan; Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
21
|
Jonasova A, Bokorova R, Polak J, Vostry M, Kostecka A, Hajkova H, Neuwirtova R, Siskova M, Sponerova D, Cermak J, Mikulenkova D, Cervinek L, Brezinova J, Michalova K, Fuchs O. High level of full-length cereblon mRNA in lower risk myelodysplastic syndrome with isolated 5q deletion is implicated in the efficacy of lenalidomide. Eur J Haematol 2014; 95:27-34. [PMID: 25284710 DOI: 10.1111/ejh.12457] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2014] [Indexed: 01/22/2023]
Abstract
Downregulation of cereblon (CRBN) gene expression is associated with resistance to the immunomodulatory drug lenalidomide and poor survival outcomes in multiple myeloma (MM) patients. However, the importance of CRBN gene expression in patients with myelodysplastic syndrome (MDS) and its impact on lenalidomide therapy are not clear. In this study, we evaluate cereblon expression in mononuclear cells isolated from bone marrow [23 lower risk MDS patients with isolated 5q deletion (5q-), 37 lower risk MDS patients with chromosome 5 without the deletion of long arms (non-5q-), and 24 healthy controls] and from peripheral blood (38 patients with 5q-, 52 non-5q- patients and 25 healthy controls) to gain insight into, firstly, the role of cereblon in lower risk MDS patients with or without 5q deletion and, secondly, into the mechanisms of lenalidomide action. Patients with 5q- lower risk MDS have the highest levels of CRBN mRNA in comparison with both lower risk MDS without the deletion of long arms of chromosome 5 and healthy controls. CRBN gene expression was measured using the quantitative TaqMan real-time PCR. High levels of CRBN mRNA were detected in all lenalidomide responders during the course of therapy. A significant decrease of the CRBN mRNA level during lenalidomide treatment is associated with loss of response to treatment and disease progression. These results suggest that, similar to the treatment of MM, high levels of full-length CRBN mRNA in lower risk 5q- patients are necessary for the efficacy of lenalidomide.
Collapse
Affiliation(s)
- Anna Jonasova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Radka Bokorova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Jaroslav Polak
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martin Vostry
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Arnost Kostecka
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Hana Hajkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Radana Neuwirtova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Magda Siskova
- 1st Medical Clinic, General University Hospital Prague, Prague, Czech Republic
| | - Dana Sponerova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jaroslav Cermak
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dana Mikulenkova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Jana Brezinova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Kyra Michalova
- Center of Oncocytogenetics, General University Hospital, Prague, Czech Republic
| | - Ota Fuchs
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
22
|
Leontieva OV, Paszkiewicz GM, Blagosklonny MV. Weekly administration of rapamycin improves survival and biomarkers in obese male mice on high-fat diet. Aging Cell 2014; 13:616-22. [PMID: 24655348 PMCID: PMC4326934 DOI: 10.1111/acel.12211] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2014] [Indexed: 01/03/2023] Open
Abstract
Recent discoveries have revealed the key role of mTOR (target of rapamycin) in aging. Furthermore, rapamycin extends lifespan in mice, especially in female mice. Here, we treated obese male mice on high-fat diet with rapamycin given intermittently: either weekly (once a week) or alternating bi-weekly (three injections every other week). While only marginally reducing obesity, intermittent administration of rapamycin significantly extended lifespan. Significance was achieved for weekly treated group and for the three rapamycin-received groups combined. In weekly treatment group, 100% mice were alive by the age of 2 years, whereas 60% of mice died in untreated group by this age. The effect of weekly treatment on survival was highly significant and cannot be fully explained by partial reduction in obesity. Alternating bi-weekly treatments seem to be less effective than weekly treatment, although effects of additional factors (see Discussion) may not be excluded. After one year of treatment, all survived mice were sacrificed 8 days after the last administration of rapamycin to avoid its direct interference with parameters examined. Fasting levels of cardiac and hepatic p-S6, a marker of mTORC1 activity, were lower in weekly treatment group compared with control mice. In contrast, levels of p-Akt (S473), glucose, triglycerides and insulin were unchanged, whereas leptin and IGF-1 tended to be lower. Thus, weekly treatment with rapamycin may slow down aging in obese male mice on high-fat diet.
Collapse
Affiliation(s)
- Olga V. Leontieva
- Cell Stress Biology Roswell Park Cancer Institute Buffalo NY 14263USA
| | | | | |
Collapse
|
23
|
Liu P, Wang Z, Wei W. Phosphorylation of Akt at the C-terminal tail triggers Akt activation. Cell Cycle 2014; 13:2162-4. [PMID: 24933731 DOI: 10.4161/cc.29584] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aberrant hyper-activation of the protein kinase Akt plays a critical role in promoting tumorigenesis. Mechanistically, previous studies establish that phosphorylation of Akt at S473 and T308 by mTORC2 and PDK1, respectively, is necessary for its full activation, thereby having been used as Akt activation markers. Recently, we report that phosphorylation of S477 and T479 at the extreme C-terminus of Akt1 promotes Akt1 activation. We further demonstrate that Akt1 pS477 and pT479 events are governed by Cdk2/Cyclin A or mTORC2 under distinct cellular contexts such as cell cycle progression or growth stimulation conditions. Here, we summarize our major findings regarding the biological significance for pS477/pT479-mediated activation of Akt and also provide perspectives for future follow-up studies.
Collapse
Affiliation(s)
- Pengda Liu
- Department of Pathology; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center; Jiangsu Institute of Hematology; The First Affiliated Hospital; Soochow University; Suzhou, PR China
| | - Wenyi Wei
- Department of Pathology; Beth Israel Deaconess Medical Center; Harvard Medical School; Boston, MA USA
| |
Collapse
|
24
|
Contact inhibition and high cell density deactivate the mammalian target of rapamycin pathway, thus suppressing the senescence program. Proc Natl Acad Sci U S A 2014; 111:8832-7. [PMID: 24889617 DOI: 10.1073/pnas.1405723111] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
During cell cycle arrest caused by contact inhibition (CI), cells do not undergo senescence, thus resuming proliferation after replating. The mechanism of senescence avoidance during CI is unknown. Recently, it was demonstrated that the senescence program, namely conversion from cell cycle arrest to senescence (i.e., geroconversion), requires mammalian target of rapamycin (mTOR). Geroconversion can be suppressed by serum starvation, rapamycin, and hypoxia, which all inhibit mTOR. Here we demonstrate that CI, as evidenced by p27 induction in normal cells, was associated with inhibition of the mTOR pathway. Furthermore, CI antagonized senescence caused by CDK inhibitors. Stimulation of mTOR in contact-inhibited cells favored senescence. In cancer cells lacking p27 induction and CI, mTOR was still inhibited in confluent culture as a result of conditioning of the medium. This inhibition of mTOR suppressed p21-induced senescence. Also, trapping of malignant cells among contact-inhibited normal cells antagonized p21-induced senescence. Thus, we identified two nonmutually exclusive mechanisms of mTOR inhibition in high cell density: (i) CI associated with p27 induction in normal cells and (ii) conditioning of the medium, especially in cancer cells. Both mechanisms can coincide in various proportions in various cells. Our work explains why CI is reversible and, most importantly, why cells avoid senescence in vivo, given that cells are contact-inhibited in the organism.
Collapse
|
25
|
Abstract
Here we discuss the latest progress in development of some kinase inhibitors such as inhibitors of c-MET, LIM and Bcr-Abl kinases. Importantly, many oncogenic kinases signal via the mTOR pathway, suggesting a common target for drug combinations.
Collapse
|