1
|
Yoon NG, Choi D, Lee JH, Kim SY, Im JY, Yun J, Yang S, Kim T, Kang S, Kang BH. Development of a Fluorescence Probe for High-Throughput Screening of Allosteric Inhibitors Targeting TRAP1. J Med Chem 2024; 67:21421-21437. [PMID: 39568139 DOI: 10.1021/acs.jmedchem.4c02343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Tumor necrosis factor receptor-associated protein 1 (TRAP1) is a molecular chaperone implicated in pro-tumorigenic pathways by regulating the folding of substrate proteins (clients) within cancer cells. Recent research has pinpointed a potentially druggable allosteric site within the client binding site (CBS) of TRAP1, suggesting this site might offer a more effective strategy for developing potent and selective TRAP1 inhibitors. However, the absence of reliable assay systems has hindered quantitative evaluation of inhibitors. In this study, we have developed a fluorescent probe, Rho6TPP, designed to target the CBS. Utilizing fluorescence polarization-based high-throughput screening assays, Rho6TPP exhibits excellent signal-to-noise ratio (>20), Z factor (>0.6), and Z' factor (>0.6). Additionally, it facilitates comparative analysis of existing small molecules and discovery of novel binders. MitoTam, a mitochondria-targeted tamoxifen, emerges as a potent CBS-targeting TRAP1 inhibitor. Our findings highlight the potential of Rho6TPP as a crucial tool for advancing the development of CBS-targeting TRAP1 inhibitors.
Collapse
Affiliation(s)
- Nam Gu Yoon
- Department of Biological Sciences, Ulsan National Institutes of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Danbi Choi
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ji Hye Lee
- Department of Biological Sciences, Ulsan National Institutes of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - So-Yeon Kim
- Department of Biological Sciences, Ulsan National Institutes of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jin Young Im
- SmartinBio Inc., Cheongju 28160, Republic of Korea
| | - Jisu Yun
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sujae Yang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Taeeun Kim
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soosung Kang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Byoung Heon Kang
- Department of Biological Sciences, Ulsan National Institutes of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- SmartinBio Inc., Cheongju 28160, Republic of Korea
| |
Collapse
|
2
|
Yang Y, Wu Y, Xiang L, Picardo M, Zhang C. Deciphering the role of skin aging in pigmentary disorders. Free Radic Biol Med 2024:S0891-5849(24)01141-9. [PMID: 39674424 DOI: 10.1016/j.freeradbiomed.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Skin aging is a complex biological process involving intrinsic and extrinsic factors. Skin aging contains alterations at the tissue, cellular, and molecular levels. Currently, there is increasing evidence that skin aging occurs not only in time-dependent chronological aging but also plays a role in skin pigmentary disorders. This review provides an in-depth analysis of the impact of skin aging on different types of pigmentary disorders, including both hyperpigmentation disorders such as melasma and senile lentigo and hypopigmentation disorders such as vitiligo, idiopathic guttate hypomelanosis and graying of hair. In addition, we explore the mechanisms of skin aging on pigmentation regulation and suggest several potential therapeutic approaches for skin aging and aging-related pigmentary disorders.
Collapse
Affiliation(s)
- Yiwen Yang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Yue Wu
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Leihong Xiang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China
| | - Mauro Picardo
- Istituto Dermopatico Immacolata, IDI-RCCS, Rome, Italy.
| | - Chengfeng Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai 200040, PR China.
| |
Collapse
|
3
|
Chen T, Bai D, Gong C, Cao Y, Yan X, Peng R. Hydrogen sulfide mitigates mitochondrial dysfunction and cellular senescence in diabetic patients: Potential therapeutic applications. Biochem Pharmacol 2024; 230:116556. [PMID: 39332692 DOI: 10.1016/j.bcp.2024.116556] [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: 08/02/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Diabetes induces a pro-aging state characterized by an increased abundance of senescent cells in various tissues, heightened chronic inflammation, reduced substance and energy metabolism, and a significant increase in intracellular reactive oxygen species (ROS) levels. This condition leads to mitochondrial dysfunction, including elevated oxidative stress, the accumulation of mitochondrial DNA (mtDNA) damage, mitophagy defects, dysregulation of mitochondrial dynamics, and abnormal energy metabolism. These dysfunctions result in intracellular calcium ion (Ca2+) homeostasis disorders, telomere shortening, immune cell damage, and exacerbated inflammation, accelerating the aging of diabetic cells or tissues. Hydrogen sulfide (H2S), a novel gaseous signaling molecule, plays a crucial role in maintaining mitochondrial function and mitigating the aging process in diabetic cells. This article systematically explores the specific mechanisms by which H2S regulates diabetes-induced mitochondrial dysfunction to delay cellular senescence, offering a promising new strategy for improving diabetes and its complications.
Collapse
Affiliation(s)
- Ting Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Dacheng Bai
- Guangdong Institute of Mitochondrial Biomedicine, Room 501, Coolpad Building, No.2 Mengxi Road, High-tech Industrial Park, Nanshan District, Shenzhen, Guangdong Province 518000, China
| | - Changyong Gong
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xiaoqing Yan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
4
|
Nunkoo VS, Cristian A, Jurcau A, Diaconu RG, Jurcau MC. The Quest for Eternal Youth: Hallmarks of Aging and Rejuvenating Therapeutic Strategies. Biomedicines 2024; 12:2540. [PMID: 39595108 PMCID: PMC11591597 DOI: 10.3390/biomedicines12112540] [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: 09/30/2024] [Revised: 10/26/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The impressive achievements made in the last century in extending the lifespan have led to a significant growth rate of elderly individuals in populations across the world and an exponential increase in the incidence of age-related conditions such as cardiovascular diseases, diabetes mellitus type 2, and neurodegenerative diseases. To date, geroscientists have identified 12 hallmarks of aging (genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, impaired macroautophagy, mitochondrial dysfunction, impaired nutrient sensing, cellular senescence, stem cell exhaustion, defective intercellular communication, chronic inflammation, and gut dysbiosis), intricately linked among each other, which can be targeted with senolytic or senomorphic drugs, as well as with more aggressive approaches such as cell-based therapies. To date, side effects seriously limit the use of these drugs. However, since rejuvenation is a dream of mankind, future research is expected to improve the tolerability of the available drugs and highlight novel strategies. In the meantime, the medical community, healthcare providers, and society should decide when to start these treatments and how to tailor them individually.
Collapse
Affiliation(s)
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
5
|
Reinema FV, Hudson N, Adema GJ, Peeters WJM, Neuzil J, Stursa J, Werner L, Sweep FCGJ, Bussink J, Span PN. MitoTam induces ferroptosis and increases radiosensitivity in head and neck cancer cells. Radiother Oncol 2024; 200:110503. [PMID: 39186982 DOI: 10.1016/j.radonc.2024.110503] [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: 03/11/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND AND PURPOSE Radiotherapy (RT) is an integral treatment part for patients with head and neck squamous cell carcinoma (HNSCC), but radioresistance remains a major issue. Here, we use MitoTam, a mitochondrially targeted analogue of tamoxifen, which we aim to stimulate ferroptotic cell death with, and sensitize radioresistant cells to RT. MATERIALS AND METHODS We assessed viability, reactive oxygen species (ROS) production, disruption of mitochondrial membrane potential, and lipid peroxidation in radiosensitive (UT-SCC-40) and radioresistant (UT-SCC-5) HNSCC cells following MitoTam treatment. To assess ferroptosis specificity, we used the ferroptosis inhibitor ferrostatin-1 (fer-1). Also, total antioxidant capacity and sensitivity to tert-butyl hydroperoxide were evaluated to assess ROS-responses. 53BP1 staining was used to assess radiosensitivity after MitoTam treatment. RESULTS Our data revealed increased ROS, cell death, disruption of mitochondrial membrane potential, and lipid peroxidation following MitoTam treatment in both cell lines. Adverse effects of MitoTam on cell death, membrane potential and lipid peroxidation were prevented by fer-1, indicating induction of ferroptosis. Radioresistant HNSCC cells were less sensitive to the effects of MitoTam due to intrinsic higher antioxidant capacity. MitoTam treatment prior to RT led to superadditive residual DNA damage expressed by 53BP1 foci compared to RT or MitoTam alone. CONCLUSION MitoTam induced ferroptosis in HNSCC cells, which could be used to overcome the elevated antioxidant capacity of radioresistant cells and sensitize such cells to RT. Treatment with MitoTam followed by RT could therefore present a promising effective therapy of radioresistant cancers. STATEMENT OF SIGNIFICANCE Radiotherapy is applied in the treatment of a majority of cancer patients. Radioresistance due to elevated antioxidant levels can be overcome by promoting ferroptotic cell death combining ROS-inducing drug MitoTam with radiotherapy.
Collapse
Affiliation(s)
- F V Reinema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - N Hudson
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - G J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - W J M Peeters
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - J Neuzil
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD 4222, Australia; Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - J Stursa
- Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - L Werner
- Faculty of Science and First Faculty of Medicine, Charles University, 120 00 Prague, Czech Republic; Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - F C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - J Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands
| | - P N Span
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen the Netherlands.
| |
Collapse
|
6
|
Ma L, Yu J, Fu Y, He X, Ge S, Jia R, Zhuang A, Yang Z, Fan X. The dual role of cellular senescence in human tumor progression and therapy. MedComm (Beijing) 2024; 5:e695. [PMID: 39161800 PMCID: PMC11331035 DOI: 10.1002/mco2.695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/21/2024] Open
Abstract
Cellular senescence, one of the hallmarks of cancer, is characterized by cell cycle arrest and the loss of most normal cellular functions while acquiring a hypersecretory, proinflammatory phenotype. The function of senescent cells in cancer cells varies depending on the cellular conditions. Before the occurrence of cancer, senescent cells act as a barrier to prevent its development. But once cancer has occurred, senescent cells play a procancer role. However, few of the current studies have adequately explained the diversity of cellular senescence across cancers. Herein, we concluded the latest intrinsic mechanisms of cellular senescence in detail and emphasized the senescence-associated secretory phenotype as a key contributor to heterogeneity of senescent cells in tumor. We also discussed five kinds of inducers of cellular senescence and the advancement of senolytics in cancer, which are drugs that tend to clear senescent cells. Finally, we summarized the various effects of senescent cells in different cancers and manifested that their functions may be diametrically opposed under different circumstances. In short, this paper contributes to the understanding of the diversity of cellular senescence in cancers and provides novel insight for tumor therapy.
Collapse
Affiliation(s)
- Liang Ma
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Jie Yu
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Yidian Fu
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Xiaoyu He
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Shengfang Ge
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Renbing Jia
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Ai Zhuang
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Zhi Yang
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| | - Xianqun Fan
- Department of OphthalmologyNinth People's HospitalShanghai JiaoTong University School of MedicineShanghaiChina
- Shanghai Key Laboratory of Orbital Diseases and Ocular OncologyShanghaiChina
| |
Collapse
|
7
|
Pan T, Yang B, Yao S, Wang R, Zhu Y. Exploring the multifaceted role of adenosine nucleotide translocase 2 in cellular and disease processes: A comprehensive review. Life Sci 2024; 351:122802. [PMID: 38857656 DOI: 10.1016/j.lfs.2024.122802] [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: 03/07/2024] [Revised: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Adenosine nucleotide translocases (ANTs) are a family of proteins abundant in the inner mitochondrial membrane, primarily responsible for shuttling ADP and ATP across the mitochondrial membrane. Additionally, ANTs are key players in balancing mitochondrial energy metabolism and regulating cell death. ANT2 isoform, highly expressed in undifferentiated and proliferating cells, is implicated in the development and drug resistance of various tumors. We conduct a detailed analysis of the potential mechanisms by which ANT2 may influence tumorigenesis and drug resistance. Notably, the significance of ANT2 extends beyond oncology, with roles in non-tumor cell processes including blood cell development, gastrointestinal motility, airway hydration, nonalcoholic fatty liver disease, obesity, chronic kidney disease, and myocardial development, making it a promising therapeutic target for multiple pathologies. To better understand the molecular mechanisms of ANT2, this review summarizes the structural properties, expression patterns, and basic functions of the ANT2 protein. In particular, we review and analyze the controversy surrounding ANT2, focusing on its role in transporting ADP/ATP across the inner mitochondrial membrane, its involvement in the composition of the mitochondrial permeability transition pore, and its participation in apoptosis.
Collapse
Affiliation(s)
- Tianhui Pan
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Bin Yang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Sheng Yao
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Rui Wang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China.
| |
Collapse
|
8
|
Qin X, Li H, Zhao H, Fang L, Wang X. Enhancing healthy aging with small molecules: A mitochondrial perspective. Med Res Rev 2024; 44:1904-1922. [PMID: 38483176 DOI: 10.1002/med.22034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/27/2024] [Accepted: 03/04/2024] [Indexed: 06/10/2024]
Abstract
The pursuit of enhanced health during aging has prompted the exploration of various strategies focused on reducing the decline associated with the aging process. A key area of this exploration is the management of mitochondrial dysfunction, a notable characteristic of aging. This review sheds light on the crucial role that small molecules play in augmenting healthy aging, particularly through influencing mitochondrial functions. Mitochondrial oxidative damage, a significant aspect of aging, can potentially be lessened through interventions such as coenzyme Q10, alpha-lipoic acid, and a variety of antioxidants. Additionally, this review discusses approaches for enhancing mitochondrial proteostasis, emphasizing the importance of mitochondrial unfolded protein response inducers like doxycycline, and agents that affect mitophagy, such as urolithin A, spermidine, trehalose, and taurine, which are vital for sustaining protein quality control. Of equal importance are methods for modulating mitochondrial energy production, which involve nicotinamide adenine dinucleotide boosters, adenosine 5'-monophosphate-activated protein kinase activators, and compounds like metformin and mitochondria-targeted tamoxifen that enhance metabolic function. Furthermore, the review delves into emerging strategies that encourage mitochondrial biogenesis. Together, these interventions present a promising avenue for addressing age-related mitochondrial degradation, thereby setting the stage for the development of innovative treatment approaches to meet this extensive challenge.
Collapse
Affiliation(s)
- Xiujiao Qin
- Department of Geriatrics, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
| | - Huiying Zhao
- Department of Geriatrics, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Le Fang
- Department of Neurology, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
- Beijing National Laboratory for Molecular Sciences, Beijing, China
| |
Collapse
|
9
|
Al Assi A, Posty S, Lamarche F, Chebel A, Guitton J, Cottet-Rousselle C, Prudent R, Lafanechère L, Giraud S, Dallemagne P, Suzanne P, Verney A, Genestier L, Castets M, Fontaine E, Billaud M, Cordier-Bussat M. A novel inhibitor of the mitochondrial respiratory complex I with uncoupling properties exerts potent antitumor activity. Cell Death Dis 2024; 15:311. [PMID: 38697987 PMCID: PMC11065874 DOI: 10.1038/s41419-024-06668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.
Collapse
Affiliation(s)
- Alaa Al Assi
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Solène Posty
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Frédéric Lamarche
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Amel Chebel
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Jérôme Guitton
- Laboratoire de biochimie et pharmacologie-toxicologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, F-69495, Pierre Bénite, France. Laboratoire de Toxicologie, Faculté de pharmacie ISPBL, Université Lyon 1, 69373, Lyon, France
| | - Cécile Cottet-Rousselle
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Renaud Prudent
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Lafanechère
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Stéphane Giraud
- Center for Drug Discovery and Development, Synergie Lyon Cancer Foundation, Lyon, Cancer Research Center, Centre Léon Bérard, Lyon, France
| | | | - Peggy Suzanne
- Normandie Univ., UNICAEN, CERMN, 14000, Caen, France
| | - Aurélie Verney
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Marie Castets
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Eric Fontaine
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France.
| | - Marc Billaud
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| | - Martine Cordier-Bussat
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| |
Collapse
|
10
|
Zhang G, Samarawickrama PN, Gui L, Ma Y, Cao M, Zhu H, Li W, Yang H, Li K, Yang Y, Zhu E, Li W, He Y. Revolutionizing Diabetic Foot Ulcer Care: The Senotherapeutic Approach. Aging Dis 2024:AD.2024.0065. [PMID: 38739931 DOI: 10.14336/ad.2024.0065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Diabetic foot ulcers (DFUs) are a prevalent and profoundly debilitating complication that afflicts individuals with diabetes mellitus (DM). These ulcers are associated with substantial morbidity, recurrence rates, disability, and mortality, imposing substantial economic, psychological, and medical burdens. Timely detection and intervention can mitigate the morbidity and disparities linked to DFU. Nevertheless, current therapeutic approaches for DFU continue to grapple with multifaceted limitations. A growing body of evidence emphasizes the crucial role of cellular senescence in the pathogenesis of chronic wounds. Interventions that try to delay cellular senescence, eliminate senescent cells (SnCs), or suppress the senescence-associated secretory phenotype (SASP) have shown promise for helping chronic wounds to heal. In this context, targeting cellular senescence emerges as a novel therapeutic strategy for DFU. In this comprehensive review, we look at the pathology and treatment of DFU in a systematic way. We also explain the growing importance of investigating SnCs in DFU and highlight the great potential of senotherapeutics that target SnCs in DFU treatment. The development of efficacious and safe senotherapeutics represents a pioneering therapeutic approach aimed at enhancing the quality of life for individuals affected by DFU.
Collapse
Affiliation(s)
- Guiqin Zhang
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| | - Li Gui
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yuan Ma
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Mei Cao
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Hong Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Wei Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Honglin Yang
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Kecheng Li
- Department of Orthopedics, the Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Yang Yang
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Enfang Zhu
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Wen Li
- Department of Endocrinology, the Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province), Kunming, Yunnan 650011, China
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650201, China
| |
Collapse
|
11
|
Kobayashi T, Fujimoto H, D’Alessandro-Gabazza CN, Yasuma T, Gabazza EC. Adenine Nucleotide Translocases and Cellular Senescence in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2024; 70:226. [PMID: 38128103 PMCID: PMC10914772 DOI: 10.1165/rcmb.2023-0386le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/20/2023] [Indexed: 12/23/2023] Open
|
12
|
Kureel SK, Blair B, Sheetz MP. Recent Advancement in Elimination Strategies and Potential Rejuvenation Targets of Senescence. Adv Biol (Weinh) 2024; 8:e2300461. [PMID: 37857532 DOI: 10.1002/adbi.202300461] [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: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Cellular senescence is a state of exiting the cell cycle, resisting apoptosis, and changing phenotype. Senescent cells (SCs) can be identified by large, distorted morphology and irreversible inability to replicate. In early development, senescence has beneficial roles like tissue patterning and wound healing, where SCs are cleared by the immune system. However, there is a steep rise in SC number as organisms age. The issue with SC accumulation stems from the loss of cellular function, alterations of the microenvironment, and secretions of pro-inflammatory molecules, consisting of cytokines, chemokines, matrix metalloproteinases (MMPs), interleukins, and extracellular matrix (ECM)-associated molecules. This secreted cocktail is referred to as the senescence-associated secretory phenotype (SASP), a hallmark of cellular senescence. The SASP promotes inflammation and displays a bystander effect where paracrine signaling turns proliferating cells into senescent states. To alleviate age-associated diseases, researchers have developed novel methods and techniques to selectively eliminate SCs in aged individuals. Although studies demonstrated that selectively killing SCs improves age-related disorders, there are drawbacks to SC removal. Considering favorable aspects of senescence in the body, this paper reviews recent advancements in elimination strategies and potential rejuvenation targets of senescence to bring researchers in the field up to date.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brandon Blair
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael P Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
13
|
Gomez LS, Jurk D. Unlocking the Potential of Senolytic Compounds: Advancements, Opportunities, and Challenges in Ageing-Related Research. Subcell Biochem 2024; 107:91-116. [PMID: 39693021 DOI: 10.1007/978-3-031-66768-8_5] [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] [Indexed: 12/19/2024]
Abstract
Cellular senescence is recognised as a contributor to the ageing process and the development of multiple age-related conditions. Researchers have launched efforts to identify compounds capable to selectively kill senescent cells, known as senolytics, without affecting non senescent cells. As of now, over 40 compounds have demonstrated senolytic properties, offering promising prospects for reversing or ameliorating age-related conditions in preclinical studies.This chapter presents the most recent developments in senolytic drug research, encompassing investigations spanning basic science, preclinical trials, and clinical studies. While many of these investigations have generated encouraging results in the realm of age-related interventions, this chapter also addresses potential challenges and pitfalls.
Collapse
Affiliation(s)
- Lilian Sales Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Woronzow V, Möhner J, Remane D, Zischler H. Generation of somatic de novo structural variation as a hallmark of cellular senescence in human lung fibroblasts. Front Cell Dev Biol 2023; 11:1274807. [PMID: 38152346 PMCID: PMC10751365 DOI: 10.3389/fcell.2023.1274807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cellular senescence is characterized by replication arrest in response to stress stimuli. Senescent cells accumulate in aging tissues and can trigger organ-specific and possibly systemic dysfunction. Although senescent cell populations are heterogeneous, a key feature is that they exhibit epigenetic changes. Epigenetic changes such as loss of repressive constitutive heterochromatin could lead to subsequent LINE-1 derepression, a phenomenon often described in the context of senescence or somatic evolution. LINE-1 elements decode the retroposition machinery and reverse transcription generates cDNA from autonomous and non-autonomous TEs that can potentially reintegrate into genomes and cause structural variants. Another feature of cellular senescence is mitochondrial dysfunction caused by mitochondrial damage. In combination with impaired mitophagy, which is characteristic of senescent cells, this could lead to cytosolic mtDNA accumulation and, as a genomic consequence, integrations of mtDNA into nuclear DNA (nDNA), resulting in mitochondrial pseudogenes called numts. Thus, both phenomena could cause structural variants in aging genomes that go beyond epigenetic changes. We therefore compared proliferating and senescent IMR-90 cells in terms of somatic de novo numts and integrations of a non-autonomous composite retrotransposons - the so-called SVA elements-that hijack the retropositional machinery of LINE-1. We applied a subtractive and kinetic enrichment technique using proliferating cell DNA as a driver and senescent genomes as a tester for the detection of nuclear flanks of de novo SVA integrations. Coupled with deep sequencing we obtained a genomic readout for SVA retrotransposition possibly linked to cellular senescence in the IMR-90 model. Furthermore, we compared the genomes of proliferative and senescent IMR-90 cells by deep sequencing or after enrichment of nuclear DNA using AluScan technology. A total of 1,695 de novo SVA integrations were detected in senescent IMR-90 cells, of which 333 were unique. Moreover, we identified a total of 81 de novo numts with perfect identity to both mtDNA and nuclear hg38 flanks. In summary, we present evidence for possible age-dependent structural genomic changes by paralogization that go beyond epigenetic modifications. We hypothesize, that the structural variants we observe potentially impact processes associated with replicative aging of IMR-90 cells.
Collapse
Affiliation(s)
- Valentina Woronzow
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jonas Möhner
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Daniel Remane
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
- HOX Life Science GmbH, Frankfurt, Hessen, Germany
| | - Hans Zischler
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
15
|
Sui J, Boatz JC, Shi J, Hu Q, Li X, Zhang Y, Königshoff M, Kliment CR. Loss of ANT1 Increases Fibrosis and Epithelial Cell Senescence in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2023; 69:556-569. [PMID: 37487137 PMCID: PMC10633847 DOI: 10.1165/rcmb.2022-0315oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 07/24/2023] [Indexed: 07/26/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is an interstitial lung disease characterized by progressive lung scarring and remodeling. Although treatments exist that slow disease progression, IPF is irreversible, and there is no cure. Cellular senescence, a major hallmark of aging, has been implicated in IPF pathogenesis, and mitochondrial dysfunction is increasingly recognized as a driver of senescence. Adenine nucleotide translocases (ANTs) are abundant mitochondrial ATP-ADP transporters critical for regulating cell fate and maintaining mitochondrial function. We sought to determine how alterations in ANTs influence cellular senescence in pulmonary fibrosis. We found that SLC25A4 (solute carrier family 25 member 4) (ANT1) and SLC25A5 (ANT2) expression is reduced in the lungs of patients with IPF, particularly within alveolar type II (AT2) cells, by single-cell RNA sequencing and tissue staining. Loss of ANT1 by siRNA in lung epithelial cells resulted in increased senescence markers such as β-galactosidase and p21, with a reduction in the ratio of nicotinamide adenine dinucleotide to reduced nicotinamide adenine dinucleotide. Bleomycin-treated ANT1 knockdown cells also had increased senescence markers compared with bleomycin-treated control cells. Loss of ANT1 in AT2 cells resulted in a reduction in alveolar organoid growth, with an increase in p21 by staining. Global loss of ANT1 resulted in worse lung fibrosis and increased senescence in the bleomycin- and asbestos-induced mouse models of pulmonary fibrosis. In summary, loss of ANT1 contributes to IPF pathogenesis through mitochondrial dysfunction, increased senescence, and decreased regenerative capacity of AT2 cells, resulting in enhanced lung fibrosis. Modulation of ANTs presents a new therapeutic avenue that may alter cellular senescence pathways and limit pulmonary fibrosis.
Collapse
Affiliation(s)
- Justin Sui
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer C Boatz
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian Shi
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Qianjiang Hu
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xiaoyun Li
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yingze Zhang
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie Königshoff
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Corrine R Kliment
- Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
16
|
Woo SH, Mo YJ, Lee YI, Park JH, Hwang D, Park TJ, Kang HY, Park SC, Lee YS. ANT2 Accelerates Cutaneous Wound Healing in Aged Skin by Regulating Energy Homeostasis and Inflammation. J Invest Dermatol 2023; 143:2295-2310.e17. [PMID: 37211200 DOI: 10.1016/j.jid.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/23/2023]
Abstract
An effective healing response is critical to healthy aging. In particular, energy homeostasis has become increasingly recognized as a factor in effective skin regeneration. ANT2 is a mediator of adenosine triphosphate import into mitochondria for energy homeostasis. Although energy homeostasis and mitochondrial integrity are critical for wound healing, the role played by ANT2 in the repair process had not been elucidated to date. In our study, we found that ANT2 expression decreased in aged skin and cellular senescence. Interestingly, overexpression of ANT2 in aged mouse skin accelerated the healing of full-thickness cutaneous wounds. In addition, upregulation of ANT2 in replicative senescent human diploid dermal fibroblasts induced their proliferation and migration, which are critical processes in wound healing. Regarding energy homeostasis, ANT2 overexpression increased the adenosine triphosphate production rate by activating glycolysis and induced mitophagy. Notably, ANT2-mediated upregulation of HSPA6 in aged human diploid dermal fibroblasts downregulated proinflammatory genes that mediate cellular senescence and mitochondrial damage. This study shows a previously uncharacterized physiological role of ANT2 in skin wound healing by regulating cell proliferation, energy homeostasis, and inflammation. Thus, our study links energy metabolism to skin homeostasis and reports, to the best of our knowledge, a previously unreported genetic factor that enhances wound healing in an aging model.
Collapse
Affiliation(s)
- Seung-Hwa Woo
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yun Jeong Mo
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Ji Hwan Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Institution of Inflamm-aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hee Young Kang
- Institution of Inflamm-aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Dermatology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sang Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju, Republic of Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea; Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
17
|
Bielcikova Z, Werner L, Stursa J, Cerny V, Krizova L, Spacek J, Hlousek S, Vocka M, Bartosova O, Pesta M, Kolostova K, Klezl P, Bobek V, Truksa J, Stemberkova-Hubackova S, Petruzelka L, Michalek P, Neuzil J. Mitochondrially targeted tamoxifen as anticancer therapy: case series of patients with renal cell carcinoma treated in a phase I/Ib clinical trial. Ther Adv Med Oncol 2023; 15:17588359231197957. [PMID: 37786538 PMCID: PMC10541747 DOI: 10.1177/17588359231197957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/03/2023] [Indexed: 10/04/2023] Open
Abstract
Mitochondrially targeted anticancer drugs (mitocans) that disrupt the energy-producing systems of cancer are emerging as new potential therapeutics. Mitochondrially targeted tamoxifen (MitoTam), an inhibitor of mitochondrial respiration respiratory complex I, is a first-in-class mitocan that was tested in the phase I/Ib MitoTam-01 trial of patients with metastatic cancer. MitoTam exhibited a manageable safety profile and efficacy; among 37% (14/38) of responders, the efficacy was greatest in patients with metastatic renal cell carcinoma (RCC) with a clinical benefit rate of 83% (5/6) of patients. This can be explained by the preferential accumulation of MitoTam in the kidney tissue in preclinical studies. Here we report the mechanism of action and safety profile of MitoTam in a case series of RCC patients. All six patients were males with a median age of 69 years, who had previously received at least three lines of palliative systemic therapy and suffered progressive disease before starting MitoTam. We recorded stable disease in four, partial response in one, and progressive disease (PD) in one patient. The histological subtype matched clear cell RCC (ccRCC) in the five responders and claro-cellular carcinoma with sarcomatoid features in the non-responder. The number of circulating tumor cells (CTCs) was evaluated longitudinally to monitor disease dynamics. Beside the decreased number of CTCs after MitoTam administration, we observed a significant decrease of the mitochondrial network mass in enriched CTCs. Two patients had long-term clinical responses to MitoTam, of 50 and 36 weeks. Both patients discontinued treatment due to adverse events, not PD. Two patients who completed the trial in November 2019 and May 2020 are still alive without subsequent anticancer therapy. The toxicity of MitoTam increased with the dosage but was manageable. The efficacy of MitoTam in pretreated ccRCC patients is linked to the novel mechanism of action of this first-in-class mitochondrially targeted drug.
Collapse
Affiliation(s)
- Zuzana Bielcikova
- Department of Oncology, General Faculty Hospital, U Nemocnice 499/2, Prague 2, 128 08, Czech Republic
| | - Lukas Werner
- Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, Prague-West 252 50, Czech Republic Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague 4, Czech Republic
| | - Jan Stursa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech RepublicDiabetes Centre, Institute for Clinical and Experimental Medicine, Prague 4, Czech Republic
| | - Vladimir Cerny
- Department of Radiodiagnostics, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Ludmila Krizova
- Department of Oncology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Spacek
- Department of Oncology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stanislav Hlousek
- Department of Oncology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michal Vocka
- Department of Oncology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Olga Bartosova
- Institute of Pharmacology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michal Pesta
- Department of Probability and Mathematical Statistics, Faculty of Mathematics and Physics, Charles University, Prague, Czech Republic
| | - Katarina Kolostova
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Petr Klezl
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic Urology Clinic, Third Faculty of Medicine, Charles University and Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Vladimir Bobek
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Sona Stemberkova-Hubackova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech RepublicDiabetes Centre, Institute for Clinical and Experimental Medicine, Prague 4, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Michalek
- Department of Anesthesiology and Intensive Care, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jiri Neuzil
- School of Pharmacy and Medical Science, Griffith University, Southport, Qld 4222, Australia Department of Pediatrics and Inherited Metabolic Diseases, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic Department of Physiology, Faculty of Science, Charles University, and General University Hospital, Prague, Czech Republic Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, Prague-West 252 50, Czech Republic
| |
Collapse
|
18
|
Kim JY, Atanassov I, Dethloff F, Kroczek L, Langer T. Time-resolved proteomic analyses of senescence highlight metabolic rewiring of mitochondria. Life Sci Alliance 2023; 6:e202302127. [PMID: 37321846 PMCID: PMC10272782 DOI: 10.26508/lsa.202302127] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
Mitochondrial dysfunction and cellular senescence are hallmarks of aging. However, the relationship between these two phenomena remains incompletely understood. In this study, we investigated the rewiring of mitochondria upon development of the senescent state in human IMR90 fibroblasts. Determining the bioenergetic activities and abundance of mitochondria, we demonstrate that senescent cells accumulate mitochondria with reduced OXPHOS activity, resulting in an overall increase of mitochondrial activities in senescent cells. Time-resolved proteomic analyses revealed extensive reprogramming of the mitochondrial proteome upon senescence development and allowed the identification of metabolic pathways that are rewired with different kinetics upon establishment of the senescent state. Among the early responding pathways, the degradation of branched-chain amino acid was increased, whereas the one carbon folate metabolism was decreased. Late-responding pathways include lipid metabolism and mitochondrial translation. These signatures were confirmed by metabolic flux analyses, highlighting metabolic rewiring as a central feature of mitochondria in cellular senescence. Together, our data provide a comprehensive view on the changes in mitochondrial proteome in senescent cells and reveal how the mitochondrial metabolism is rewired in senescent cells.
Collapse
Affiliation(s)
- Jun Yong Kim
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ilian Atanassov
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Lara Kroczek
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Nehlin JO. Senolytic and senomorphic interventions to defy senescence-associated mitochondrial dysfunction. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:217-247. [PMID: 37437979 DOI: 10.1016/bs.apcsb.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The accumulation of senescent cells in the aging individual is associated with an increase in the occurrence of age-associated pathologies that contribute to poor health, frailty, and mortality. The number and type of senescent cells is viewed as a contributor to the body's senescence burden. Cellular models of senescence are based on induction of senescence in cultured cells in the laboratory. One type of senescence is triggered by mitochondrial dysfunction. There are several indications that mitochondria defects contribute to body aging. Senotherapeutics, targeting senescent cells, have been shown to induce their lysis by means of senolytics, or repress expression of their secretome, by means of senomorphics, senostatics or gerosuppressors. An outline of the mechanism of action of various senotherapeutics targeting mitochondria and senescence-associated mitochondria dysfunction will be here addressed. The combination of geroprotective interventions together with senotherapeutics will help to strengthen mitochondrial energy metabolism, biogenesis and turnover, and lengthen the mitochondria healthspan, minimizing one of several molecular pathways contributing to the aging phenotype.
Collapse
Affiliation(s)
- Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark.
| |
Collapse
|
20
|
Liao KM, Chen CJ, Luo WJ, Hsu CW, Yu SL, Yang PC, Su KY. Senomorphic effect of diphenyleneiodonium through AMPK/MFF/DRP1 mediated mitochondrial fission. Biomed Pharmacother 2023; 162:114616. [PMID: 37004322 DOI: 10.1016/j.biopha.2023.114616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
With an aging population and the numerous health impacts associated with old age, the identification of anti-aging drugs has become an important new research direction. Although mitochondria have been recognized to affect aging, anti-aging drugs specifically targeting the mitochondria are less well characterized. In this study, diphenyleneiodonium (DPI) was identified as a potential senomorphic drug that functions by promoting mitochondrial fission. DPI significantly reduced the number of senescence-associated β-galactosidase (SA-β-gal) positive cells and increased the number of proliferating Ki-67 positive cells in BrdU or irradiation stress-induced senescent NIH3T3 cells or IMR90 cells and mouse embryonic fibroblasts (MEFs) replicative senescent cells. Cell cycle arrest genes and senescence-associated secretory phenotype (SASP) factors were downregulated with DPI treatment. In addition, the oxygen consumption rate (OCR) of mitochondrial respiration showed that DPI significantly reduced senescence-associated hyper OCR. Mechanistically, DPI promoted mitochondrial fission by enhancing AMPK/MFF phosphorylation and DRP1 mitochondrial translocation. Inhibition of DRP1 by Mdivi-1 abolished DPI-induced mitochondrial fission and the anti-senescence phenotype. Importantly, Eighty-eight-week-old mice treated with DPI had significantly reduced numbers of SA-β-gal positive cells and reduced expression of cell cycle arrest genes and SASP factors in their livers and kidneys. Pathological and functional assays showed DPI treatment not only reduced liver fibrosis and immune cell infiltration but also improved aged-related physical impairments in aged mice. Taken together, our study identified a potential anti-aging compound that exerts its effects through modulation of mitochondrial morphology.
Collapse
|
21
|
Khalil R, Diab-Assaf M, Lemaitre JM. Emerging Therapeutic Approaches to Target the Dark Side of Senescent Cells: New Hopes to Treat Aging as a Disease and to Delay Age-Related Pathologies. Cells 2023; 12:915. [PMID: 36980256 PMCID: PMC10047596 DOI: 10.3390/cells12060915] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Life expectancy has drastically increased over the last few decades worldwide, with important social and medical burdens and costs. To stay healthy longer and to avoid chronic disease have become essential issues. Organismal aging is a complex process that involves progressive destruction of tissue functionality and loss of regenerative capacity. One of the most important aging hallmarks is cellular senescence, which is a stable state of cell cycle arrest that occurs in response to cumulated cell stresses and damages. Cellular senescence is a physiological mechanism that has both beneficial and detrimental consequences. Senescence limits tumorigenesis, lifelong tissue damage, and is involved in different biological processes, such as morphogenesis, regeneration, and wound healing. However, in the elderly, senescent cells increasingly accumulate in several organs and secrete a combination of senescence associated factors, contributing to the development of various age-related diseases, including cancer. Several studies have revealed major molecular pathways controlling the senescent phenotype, as well as the ones regulating its interactions with the immune system. Attenuating the senescence-associated secretory phenotype (SASP) or eliminating senescent cells have emerged as attractive strategies aiming to reverse or delay the onset of aging diseases. Here, we review current senotherapies designed to suppress the deleterious effect of SASP by senomorphics or to selectively kill senescent cells by "senolytics" or by immune system-based approaches. These recent investigations are promising as radical new controls of aging pathologies and associated multimorbidities.
Collapse
Affiliation(s)
- Roula Khalil
- IRMB, University Montpellier, INSERM, 34090 Montpellier, France;
| | - Mona Diab-Assaf
- Fanar Faculty of Sciences II, Lebanese University, Beirut P.O. Box 90656, Lebanon;
| | | |
Collapse
|
22
|
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS J 2023; 290:1362-1383. [PMID: 35015337 DOI: 10.1111/febs.16350] [Citation(s) in RCA: 200] [Impact Index Per Article: 200.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/17/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022]
Abstract
The concept of geroscience is that since ageing is the greatest risk factor for many diseases and conditions, targeting the ageing process itself will have the greatest impact on human health. Of the hallmarks of ageing, cellular senescence has emerged as a druggable therapeutic target for extending healthspan in model organisms. Cellular senescence is a cell state of irreversible proliferative arrest driven by different types of stress, including oncogene-induced stress. Many senescent cells (SnCs) develop a senescent-associated secretory phenotype (SASP) comprising pro-inflammatory cytokines, chemokines, proteases, bioactive lipids, inhibitory molecules, extracellular vesicles, metabolites, lipids and other factors, able to promote chronic inflammation and tissue dysfunction. SnCs up-regulate senescent cell anti-apoptotic pathways (SCAPs) that prevent them from dying despite the accumulation of damage to DNA and other organelles. These SCAPs and other pathways altered in SnCs represent therapeutic targets for the development of senotherapeutic drugs that induce selective cell death of SnCs, specifically termed senolytics or suppress markers of senescence, in particular the SASP, termed senomorphics. Here, we review the current state of the development of senolytics and senomorphics for the treatment of age-related diseases and disorders and extension of healthy longevity. In addition, the challenges of documenting senolytic and senomorphic activity in pre-clinical models and the current state of the clinical application of the different senotherapeutics will be discussed.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Louise E Pitcher
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Vaishali Prahalad
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
23
|
Martini H, Passos JF. Cellular senescence: all roads lead to mitochondria. FEBS J 2023; 290:1186-1202. [PMID: 35048548 PMCID: PMC9296701 DOI: 10.1111/febs.16361] [Citation(s) in RCA: 109] [Impact Index Per Article: 109.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 01/10/2023]
Abstract
Senescence is a multi-functional cell fate, characterized by an irreversible cell-cycle arrest and a pro-inflammatory phenotype, commonly known as the senescence-associated secretory phenotype (SASP). Emerging evidence indicates that accumulation of senescent cells in multiple tissues drives tissue dysfunction and several age-related conditions. This has spurred the academic community and industry to identify new therapeutic interventions targeting this process. Mitochondrial dysfunction is an often-unappreciated hallmark of cellular senescence which plays important roles not only in the senescence growth arrest but also in the development of the SASP and resistance to cell-death. Here, we review the evidence that supports a role for mitochondria in the development of senescence and describe the underlying mechanisms. Finally, we propose that a detailed road map of mitochondrial biology in senescence will be crucial to guide the future development of senotherapies.
Collapse
Affiliation(s)
- Hélène Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905 USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905 USA
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905 USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905 USA
| |
Collapse
|
24
|
Bielcikova Z, Stursa J, Krizova L, Dong L, Spacek J, Hlousek S, Vocka M, Rohlenova K, Bartosova O, Cerny V, Padrta T, Pesta M, Michalek P, Hubackova SS, Kolostova K, Pospisilova E, Bobek V, Klezl P, Zobalova R, Endaya B, Rohlena J, Petruzelka L, Werner L, Neuzil J. Mitochondrially targeted tamoxifen in patients with metastatic solid tumours: an open-label, phase I/Ib single-centre trial. EClinicalMedicine 2023; 57:101873. [PMID: 37064512 PMCID: PMC10102891 DOI: 10.1016/j.eclinm.2023.101873] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/10/2023] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
Background Mitochondria present an emerging target for cancer treatment. We have investigated the effect of mitochondrially targeted tamoxifen (MitoTam), a first-in-class anti-cancer agent, in patients with solid metastatic tumours. Methods MitoTam was tested in an open-label, single-centre (Department of Oncology, General Faculty Hospital, Charles University, Czech Republic), phase I/Ib trial in metastatic patients with various malignancies and terminated oncological therapies. In total, 75 patients were enrolled between May 23, 2018 and July 22, 2020. Phase I evaluated escalating doses of MitoTam in two therapeutic regimens using the 3 + 3 design to establish drug safety and maximum tolerated dose (MTD). In phase Ib, three dosing regimens were applied over 8 and 6 weeks to evaluate long-term toxicity of MitoTam as the primary objective and its anti-cancer effect as a secondary objective. This trial was registered with the European Medicines Agency under EudraCT 2017-004441-25. Findings In total, 37 patients were enrolled into phase I and 38 into phase Ib. In phase I, the initial application of MitoTam via peripheral vein indicated high risk of thrombophlebitis, which was avoided by central vein administration. The highest dose with acceptable side effects was 5.0 mg/kg. The prevailing adverse effects (AEs) in phase I were neutropenia (30%), anaemia (30%) and fever/hyperthermia (30%), and in phase Ib fever/hyperthermia (58%) together with anaemia (26%) and neutropenia (16%). Serious AEs were mostly related to thromboembolic (TE) complications that affected 5% and 13% of patients in phase I and Ib, respectively. The only statistically significant AE related to MitoTam treatment was anaemia in phase Ib (p = 0.004). Of the tested regimens weekly dosing with 3.0 mg/kg for 6 weeks afforded the best safety profile with almost all being grade 1 (G1) AEs. Altogether, five fatalities occurred during the study, two of them meeting criteria for Suspected Unexpected Serious Adverse Events Reporting (SUSAR) (G4 thrombocytopenia and G5 stroke). MitoTam showed benefit evaluated as clinical benefit rate (CBR) in 37% patients with the largest effect in renal cell carcinoma (RCC) where four out of six patients reached disease stabilisation (SD), one reached partial response (PR) so that in total, five out of six (83%) patients showed CBR. Interpretation In this study, the MTD was established as 5.0 mg/kg and the recommended dose of MitoTam as 3.0 mg/kg given once per week via central vein with recommended preventive anti-coagulation therapy. The prevailing toxicity included haematological AEs, hyperthermia/fever and TE complications. One fatal stroke and non-fatal G4 thrombocytopenia were recorded. MitoTam showed high efficacy against RCC. Funding Smart Brain Ltd. Translation For the Czech translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Zuzana Bielcikova
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
- Corresponding author. Department of Oncology, General Faculty Hospital and 1st Faculty of Medicine, Charles University, U Nemocnice 499/2, Prague 2 128 08, Czech Republic.
| | - Jan Stursa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - Ludmila Krizova
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Lanfeng Dong
- School of Pharmacy and Medical Science, Griffith University, Southport, Qld 4222, Australia
| | - Jan Spacek
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Stanislav Hlousek
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Katerina Rohlenova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - Olga Bartosova
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague 128 08, Czech Republic
| | - Vladimir Cerny
- Department of Radiodiagnostics, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Tomas Padrta
- Department of Radiodiagnostics, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Michal Pesta
- Department of Probability and Mathematical Statistics, Faculty of Mathematics and Physics, Charles University, Prague 121 06, Czech Republic
| | - Pavel Michalek
- Department of Anesthesiology and Intensive Care, First Faculty of Medicine, Charles University and General University Hospital, Prague 128 08, Czech Republic
| | - Sona Stemberkova Hubackova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague 4 140 21, Czech Republic
| | - Katarina Kolostova
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague 10 100 34, Czech Republic
| | - Eliska Pospisilova
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague 10 100 34, Czech Republic
| | - Vladimir Bobek
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague 10 100 34, Czech Republic
| | - Peter Klezl
- Laboratory of Personalized Medicine, Oncology Clinic, Faculty Hospital Kralovske Vinohrady, Prague 10 100 34, Czech Republic
- Urology Clinic, Third Faculty of Medicine, Charles University and Faculty Hospital Kralovske Vinohrady, Prague 10 100 34, Czech Republic
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - Berwini Endaya
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
- Department of Pediatrics and Inherited Metabolic Diseases, First Faculty of Medicine, Charles University, Prague 2 128 08, Czech Republic
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague 128 08, Czech Republic
| | - Lukas Werner
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
- Corresponding author. Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, Prague-West 252 50, Czech Republic.
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West 252 50, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, Qld 4222, Australia
- Department of Pediatrics and Inherited Metabolic Diseases, First Faculty of Medicine, Charles University, Prague 2 128 08, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague 2 128 00, Czech Republic
- Corresponding author. School of Pharmacy and Medical Science, Griffith University, Parklands Avenue, 4222 Southport, Qld, Australia, or Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, Prague-West 252 50, Czech Republic.
| |
Collapse
|
25
|
Shi Y, Luo Z, You J. Subcellular delivery of lipid nanoparticles to endoplasmic reticulum and mitochondria. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1803. [PMID: 35441489 DOI: 10.1002/wnan.1803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 06/14/2023]
Abstract
Primarily responsible for the biogenesis and metabolism of biomolecules, endoplasmic reticulum (ER) and mitochondria are gradually becoming the targets of therapeutic modulation, whose physiological activities and pathological manifestations determine the functional capacity and even the survival of cells. Drug delivery systems with specific physicochemical properties (passive targeting), or modified by small molecular compounds, polypeptides, and biomembranes demonstrating tropism for ER and mitochondria (active targeting) are able to reduce the nonselective accumulation of drugs, enhancing efficacy while reducing side effects. Lipid nanoparticles feature high biocompatibility, diverse cargo loading, and flexible structure modification, which are frequently used for subcellular organelle-targeted delivery of therapeutics. However, there is still a lack of systematic understanding of lipid nanoparticle-based ER and mitochondria targeting. Herein, we review the pathological significance of drug selectively delivered to the ER and mitochondria. We also summarize the molecular basis and application prospects of lipid nanoparticle-based ER and mitochondria targeting strategies, which may provide guidance for the prevention and treatment of associated diseases and disorders. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Arbon D, Ženíšková K, Šubrtová K, Mach J, Štursa J, Machado M, Zahedifard F, Leštinová T, Hierro-Yap C, Neuzil J, Volf P, Ganter M, Zoltner M, Zíková A, Werner L, Sutak R. Repurposing of MitoTam: Novel Anti-Cancer Drug Candidate Exhibits Potent Activity against Major Protozoan and Fungal Pathogens. Antimicrob Agents Chemother 2022; 66:e0072722. [PMID: 35856666 PMCID: PMC9380531 DOI: 10.1128/aac.00727-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/20/2022] Open
Abstract
Many of the currently available anti-parasitic and anti-fungal frontline drugs have severe limitations, including adverse side effects, complex administration, and increasing occurrence of resistance. The discovery and development of new therapeutic agents is a costly and lengthy process. Therefore, repurposing drugs with already established clinical application offers an attractive, fast-track approach for novel treatment options. In this study, we show that the anti-cancer drug candidate MitoTam, a mitochondria-targeted analog of tamoxifen, efficiently eliminates a wide range of evolutionarily distinct pathogens in vitro, including pathogenic fungi, Plasmodium falciparum, and several species of trypanosomatid parasites, causative agents of debilitating neglected tropical diseases. MitoTam treatment was also effective in vivo and significantly reduced parasitemia of two medically important parasites, Leishmania mexicana and Trypanosoma brucei, in their respective animal infection models. Functional analysis in the bloodstream form of T. brucei showed that MitoTam rapidly altered mitochondrial functions, particularly affecting cellular respiration, lowering ATP levels, and dissipating mitochondrial membrane potential. Our data suggest that the mode of action of MitoTam involves disruption of the inner mitochondrial membrane, leading to rapid organelle depolarization and cell death. Altogether, MitoTam is an excellent candidate drug against several important pathogens, for which there are no efficient therapies and for which drug development is not a priority.
Collapse
Affiliation(s)
- Dominik Arbon
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Kateřina Ženíšková
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Karolína Šubrtová
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Mach
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Štursa
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Marta Machado
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Farnaz Zahedifard
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Tereza Leštinová
- Faculty of Sciences, Charles University, Department of Parasitology, Prague, Czech Republic
| | - Carolina Hierro-Yap
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
- School of Pharmacy and Medical Science, Griffith University, Southport, Queensland, Australia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Faculty of Sciences, Charles University, Department of Parasitology, Prague, Czech Republic
| | - Markus Ganter
- Centre for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Zoltner
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Alena Zíková
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lukáš Werner
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Robert Sutak
- Department of Parasitology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
27
|
Zhang L, Liu Y, Zhou R, He B, Wang W, Zhang B. Cyclophilin D: Guardian or Executioner for Tumor Cells? Front Oncol 2022; 12:939588. [PMID: 35860554 PMCID: PMC9289278 DOI: 10.3389/fonc.2022.939588] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclophilin D (CypD) is a peptide-proline cis-trans isomerase (PPIase) distributed in the mitochondrial matrix. CypD regulates the opening of the mitochondrial permeability conversion pore (mPTP) and mitochondrial bioenergetics through PPIase activity or interaction with multiple binding partners in mitochondria. CypD initially attracted attention due to its regulation of mPTP overopening-mediated cell death. However, recent studies on the effects of CypD on tumors have shown conflicting results. Although CypD has been proven to promote the aerobic glycolysis in tumor cells, its regulation of malignant characteristics such as the survival, invasion and drug resistance of tumor cells remains controversial. Here, we elaborate the main biological functions of CypD and its relationships with tumor progression identified in recent years, focusing on the dual role of CypD in tumors.
Collapse
Affiliation(s)
- Ling Zhang
- School of Nursing, Jining Medical University, Jining, China
- *Correspondence: Bin Zhang, ; Ling Zhang,
| | - Yi Liu
- School of Nursing, Jining Medical University, Jining, China
- School of Public Health, North China University of Science and Technology, Tangshan, China
| | - Rou Zhou
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Wenjun Wang
- School of Nursing, Jining Medical University, Jining, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
- *Correspondence: Bin Zhang, ; Ling Zhang,
| |
Collapse
|
28
|
Aghali A, Koloko Ngassie ML, Pabelick CM, Prakash YS. Cellular Senescence in Aging Lungs and Diseases. Cells 2022; 11:cells11111781. [PMID: 35681476 PMCID: PMC9179897 DOI: 10.3390/cells11111781] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Cellular senescence represents a state of irreversible cell cycle arrest occurring naturally or in response to exogenous stressors. Following the initial arrest, progressive phenotypic changes define conditions of cellular senescence. Understanding molecular mechanisms that drive senescence can help to recognize the importance of such pathways in lung health and disease. There is increasing interest in the role of cellular senescence in conditions such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) in the context of understanding pathophysiology and identification of novel therapies. Herein, we discuss the current knowledge of molecular mechanisms and mitochondrial dysfunction regulating different aspects of cellular senescence-related to chronic lung diseases to develop rational strategies for modulating the senescent cell phenotype in the lung for therapeutic benefit.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
| | - Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Christina M. Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| |
Collapse
|
29
|
Kudlova N, De Sanctis JB, Hajduch M. Cellular Senescence: Molecular Targets, Biomarkers, and Senolytic Drugs. Int J Mol Sci 2022; 23:ijms23084168. [PMID: 35456986 PMCID: PMC9028163 DOI: 10.3390/ijms23084168] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is defined as irreversible cell cycle arrest caused by various processes that render viable cells non-functional, hampering normal tissue homeostasis. It has many endogenous and exogenous inducers, and is closely connected with age, age-related pathologies, DNA damage, degenerative disorders, tumor suppression and activation, wound healing, and tissue repair. However, the literature is replete with contradictory findings concerning its triggering mechanisms, specific biomarkers, and detection protocols. This may be partly due to the wide range of cellular and in vivo animal or human models of accelerated aging that have been used to study senescence and test senolytic drugs. This review summarizes recent findings concerning senescence, presents some widely used cellular and animal senescence models, and briefly describes the best-known senolytic agents.
Collapse
Affiliation(s)
- Natalie Kudlova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine Czech Advanced Technologies and Research Institute, Palacky University, 77147 Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine Czech Advanced Technologies and Research Institute, Palacky University, 77147 Olomouc, Czech Republic
- Correspondence: ; Tel.: +42-0-585632082
| |
Collapse
|
30
|
Vacurova E, Trnovska J, Svoboda P, Skop V, Novosadova V, Reguera DP, Petrezselyová S, Piavaux B, Endaya B, Spoutil F, Zudova D, Stursa J, Melcova M, Bielcikova Z, Werner L, Prochazka J, Sedlacek R, Huttl M, Hubackova SS, Haluzik M, Neuzil J. Mitochondrially targeted tamoxifen alleviates markers of obesity and type 2 diabetes mellitus in mice. Nat Commun 2022; 13:1866. [PMID: 35387987 PMCID: PMC8987092 DOI: 10.1038/s41467-022-29486-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/17/2022] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus represents a major health problem with increasing prevalence worldwide. Limited efficacy of current therapies has prompted a search for novel therapeutic options. Here we show that treatment of pre-diabetic mice with mitochondrially targeted tamoxifen, a potential anti-cancer agent with senolytic activity, improves glucose tolerance and reduces body weight with most pronounced reduction of visceral adipose tissue due to reduced food intake, suppressed adipogenesis and elimination of senescent cells. Glucose-lowering effect of mitochondrially targeted tamoxifen is linked to improvement of type 2 diabetes mellitus-related hormones profile and is accompanied by reduced lipid accumulation in liver. Lower senescent cell burden in various tissues, as well as its inhibitory effect on pre-adipocyte differentiation, results in lower level of circulating inflammatory mediators that typically enhance metabolic dysfunction. Targeting senescence with mitochodrially targeted tamoxifen thus represents an approach to the treatment of type 2 diabetes mellitus and its related comorbidities, promising a complex impact on senescence-related pathologies in aging population of patients with type 2 diabetes mellitus with potential translation into the clinic.
Collapse
Affiliation(s)
- Eliska Vacurova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Jaroslava Trnovska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petr Svoboda
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Vojtech Skop
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, 20892, USA
| | - Vendula Novosadova
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - David Pajuelo Reguera
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Silvia Petrezselyová
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Benoit Piavaux
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Berwini Endaya
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Frantisek Spoutil
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Dagmar Zudova
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jan Stursa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Magdalena Melcova
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Prague, Czech Republic
| | | | - Lukas Werner
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jan Prochazka
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Radislav Sedlacek
- Institute of Molecular Genetics, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Martina Huttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic.
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia.
| |
Collapse
|
31
|
Hönigova K, Navratil J, Peltanova B, Polanska HH, Raudenska M, Masarik M. Metabolic tricks of cancer cells. Biochim Biophys Acta Rev Cancer 2022; 1877:188705. [PMID: 35276232 DOI: 10.1016/j.bbcan.2022.188705] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/11/2022] [Accepted: 02/26/2022] [Indexed: 12/15/2022]
Abstract
One of the characteristics of cancer cells important for tumorigenesis is their metabolic plasticity. Indeed, in various stress conditions, cancer cells can reshape their metabolic pathways to support the increased energy request due to continuous growth and rapid proliferation. Moreover, selective pressures in the tumor microenvironment, such as hypoxia, acidosis, and competition for resources, force cancer cells to adapt by complete reorganization of their metabolism. In this review, we highlight the characteristics of cancer metabolism and discuss its clinical significance, since overcoming metabolic plasticity of cancer cells is a key objective of modern cancer therapeutics and a better understanding of metabolic reprogramming may lead to the identification of possible targets for cancer therapy.
Collapse
Affiliation(s)
- Katerina Hönigova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Jiri Navratil
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Barbora Peltanova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Hana Holcova Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic; BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, CZ-252 50 Vestec, Czech Republic.
| |
Collapse
|
32
|
Hong X, Wang L, Zhang K, Liu J, Liu JP. Molecular Mechanisms of Alveolar Epithelial Stem Cell Senescence and Senescence-Associated Differentiation Disorders in Pulmonary Fibrosis. Cells 2022; 11:877. [PMID: 35269498 PMCID: PMC8909789 DOI: 10.3390/cells11050877] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary senescence is accelerated by unresolved DNA damage response, underpinning susceptibility to pulmonary fibrosis. Recently it was reported that the SARS-Cov-2 viral infection induces acute pulmonary epithelial senescence followed by fibrosis, although the mechanism remains unclear. Here, we examine roles of alveolar epithelial stem cell senescence and senescence-associated differentiation disorders in pulmonary fibrosis, exploring the mechanisms mediating and preventing pulmonary fibrogenic crisis. Notably, the TGF-β signalling pathway mediates alveolar epithelial stem cell senescence by mechanisms involving suppression of the telomerase reverse transcriptase gene in pulmonary fibrosis. Alternatively, telomere uncapping caused by stress-induced telomeric shelterin protein TPP1 degradation mediates DNA damage response, pulmonary senescence and fibrosis. However, targeted intervention of cellular senescence disrupts pulmonary remodelling and fibrosis by clearing senescent cells using senolytics or preventing senescence using telomere dysfunction inhibitor (TELODIN). Studies indicate that the development of senescence-associated differentiation disorders is reprogrammable and reversible by inhibiting stem cell replicative senescence in pulmonary fibrosis, providing a framework for targeted intervention of the molecular mechanisms of alveolar stem cell senescence and pulmonary fibrosis. Abbreviations: DPS, developmental programmed senescence; IPF, idiopathic pulmonary fibrosis; OIS, oncogene-induced replicative senescence; SADD, senescence-associated differentiation disorder; SALI, senescence-associated low-grade inflammation; SIPS, stress-induced premature senescence; TERC, telomerase RNA component; TERT, telomerase reverse transcriptase; TIFs, telomere dysfunction-induced foci; TIS, therapy-induced senescence; VIS, virus-induced senescence.
Collapse
Affiliation(s)
- Xiaojing Hong
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Kexiong Zhang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, VIC 3181, Australia
- Hudson Institute of Medical Research, Monash University Department of Molecular and Translational Science, Clayton, VIC 3168, Australia
| |
Collapse
|
33
|
Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: Eliminating Senescent Cells and Alleviating Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2022; 10:823945. [PMID: 35309994 PMCID: PMC8924288 DOI: 10.3389/fbioe.2022.823945] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is the main cause of cervical and lumbar spondylosis. Over the past few years, the relevance between cellular senescence and IVDD has been widely studied, and the senescence-associated secretory phenotype (SASP) produced by senescent cells is found to remodel extracellular matrix (ECM) metabolism and destruct homeostasis. Elimination of senescent cells by senolytics and suppression of SASP production by senomorphics/senostatics are effective strategies to alleviate degenerative diseases including IVDD. Here, we review the involvement of senescence in the process of IVDD; we also discuss the potential of senolytics on eliminating senescent disc cells and alleviating IVDD; finally, we provide a table listing senolytic drugs and small molecules, aiming to propose potential drugs for IVDD therapy in the future.
Collapse
Affiliation(s)
- Yuhao Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shiwei Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| |
Collapse
|
34
|
Morsli S, Doherty GJ, Muñoz-Espín D. Activatable senoprobes and senolytics: Novel strategies to detect and target senescent cells. Mech Ageing Dev 2022; 202:111618. [PMID: 34990647 DOI: 10.1016/j.mad.2021.111618] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/21/2021] [Accepted: 12/24/2021] [Indexed: 01/10/2023]
Abstract
Pharmacologically active compounds that manipulate cellular senescence (senotherapies) have recently shown great promise in multiple pre-clinical disease models, and some of them are now being tested in clinical trials. Despite promising proof-of-principle evidence, there are known on- and off-target toxicities associated with these compounds, and therefore more refined and novel strategies to improve their efficacy and specificity for senescent cells are being developed. Preferential release of drugs and macromolecular formulations within senescent cells has been predominantly achieved by exploiting one of the most widely used biomarkers of senescence, the increase in lysosomal senescence-associated β-galactosidase (SA-β-gal) activity, a common feature of most reported senescent cell types. Galacto-conjugation is a versatile therapeutic and detection strategy to facilitate preferential targeting of senescent cells by using a variety of existing formulations, including modular systems, nanocarriers, activatable prodrugs, probes, and small molecules. We discuss the benefits and drawbacks of these specific senescence targeting tools and how the strategy of galacto-conjugation might be utilised to design more specific and sophisticated next-generation senotherapeutics, as well as theranostic agents. Finally, we discuss some innovative strategies and possible future directions for the field.
Collapse
Affiliation(s)
- Samir Morsli
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK
| | - Gary J Doherty
- Department of Oncology, Box 193, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK.
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| |
Collapse
|
35
|
Chiang JC, Chen WM, Newman C, Chen BPC, Lee H. Lysophosphatidic Acid Receptor 3 Promotes Mitochondrial Homeostasis against Oxidative Stress: Potential Therapeutic Approaches for Hutchinson–Gilford Progeria Syndrome. Antioxidants (Basel) 2022; 11:antiox11020351. [PMID: 35204233 PMCID: PMC8869156 DOI: 10.3390/antiox11020351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a growth factor-like lipid mediator that regulates various physiological functions via activation of multiple LPA G protein-coupled receptors. We previously reported that LPA suppresses oxidative stress in premature aging Hutchinson-Gilford progeria syndrome (HGPS) patient fibroblasts via its type 3 receptor (LPA3). Mitochondria have been suggested to be the primary origin of oxidative stress via the overproduction of reactive oxygen species (ROS). Mitochondria are responsible for producing ATP through oxidative phosphorylation (OXPHOS) and have a calcium buffering capacity for the cell. Defects in mitochondria will lead to declined antioxidant capacity and cell apoptosis. Therefore, we aim to demonstrate the regulatory role of LPA3 in mitochondrial homeostasis. siRNA-mediated depletion of LPA3 leads to the depolarization of mitochondrial potential (ΔΨm) and cellular ROS accumulation. In addition, the depletion of LPA3 enhances cisplatin-induced cytochrome C releasing. This indicates that LPA3 is essential to suppress the mitochondrial apoptosis pathway. LPA3 is also shown to improve mitochondrial ADP-ATP exchange by enhancing the protein level of ANT2. On the other hand, LPA3 regulates calcium uptake from the ER to mitochondria via the IP3R1-VDAC1 channel. Moreover, activation of LPA3 by selective agonist OMPT rescues mitochondrial homeostasis of H2O2-induced oxidative stress cells and HGPS patient fibroblasts by improving mitochondrial ΔΨm and OXPHOS. In summary, our findings imply that LPA3 acts as the gatekeeper for mitochondrial healthiness to maintain cell youth. Furthermore, LPA3 can be a promising therapeutic target to prevent mitochondrial oxidative stress in aging and HGPS.
Collapse
Affiliation(s)
- Jui-Chung Chiang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (J.-C.C.); (W.-M.C.); (C.N.)
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Wei-Min Chen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (J.-C.C.); (W.-M.C.); (C.N.)
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Ciara Newman
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (J.-C.C.); (W.-M.C.); (C.N.)
| | - Benjamin P. C. Chen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (J.-C.C.); (W.-M.C.); (C.N.)
- Correspondence: (B.P.C.C.); (H.L.); Tel.: +1-214-648-1263 (B.P.C.C.); +886-2-3366-2499 (H.L.)
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: (B.P.C.C.); (H.L.); Tel.: +1-214-648-1263 (B.P.C.C.); +886-2-3366-2499 (H.L.)
| |
Collapse
|
36
|
Le HH, Cinaroglu SS, Manalo EC, Ors A, Gomes MM, Duan Sahbaz B, Bonic K, Origel Marmolejo CA, Quentel A, Plaut JS, Kawashima TE, Ozdemir ES, Malhotra SV, Ahiska Y, Sezerman U, Bayram Akcapinar G, Saldivar JC, Timucin E, Fischer JM. Molecular modelling of the FOXO4-TP53 interaction to design senolytic peptides for the elimination of senescent cancer cells. EBioMedicine 2021; 73:103646. [PMID: 34689087 PMCID: PMC8546421 DOI: 10.1016/j.ebiom.2021.103646] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Senescent cells accumulate in tissues over time as part of the natural ageing process and the removal of senescent cells has shown promise for alleviating many different age-related diseases in mice. Cancer is an age-associated disease and there are numerous mechanisms driving cellular senescence in cancer that can be detrimental to recovery. Thus, it would be beneficial to develop a senolytic that acts not only on ageing cells but also senescent cancer cells to prevent cancer recurrence or progression. METHODS We used molecular modelling to develop a series of rationally designed peptides to mimic and target FOXO4 disrupting the FOXO4-TP53 interaction and releasing TP53 to induce apoptosis. We then tested these peptides as senolytic agents for the elimination of senescent cells both in cell culture and in vivo. FINDINGS Here we show that these peptides can act as senolytics for eliminating senescent human cancer cells both in cell culture and in orthotopic mouse models. We then further characterized one peptide, ES2, showing that it disrupts FOXO4-TP53 foci, activates TP53 mediated apoptosis and preferentially binds FOXO4 compared to TP53. Next, we show that intratumoural delivery of ES2 plus a BRAF inhibitor results in a significant increase in apoptosis and a survival advantage in mouse models of melanoma. Finally, we show that repeated systemic delivery of ES2 to older mice results in reduced senescent cell numbers in the liver with minimal toxicity. INTERPRETATION Taken together, our results reveal that peptides can be generated to specifically target and eliminate FOXO4+ senescent cancer cells, which has implications for eradicating residual disease and as a combination therapy for frontline treatment of cancer. FUNDING This work was supported by the Cancer Early Detection Advanced Research Center at Oregon Health & Science University.
Collapse
Affiliation(s)
- Hillary H Le
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Suleyman S Cinaroglu
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir Istanbul 34752, Turkey; Eternans Ltd., UK
| | - Elise C Manalo
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Aysegul Ors
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Michelle M Gomes
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | | | - Karla Bonic
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Carlos A Origel Marmolejo
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Arnaud Quentel
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Justin S Plaut
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA; Dept of Bioengineering, University of California San Diego, USA
| | - Taryn E Kawashima
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - E Sila Ozdemir
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Sanjay V Malhotra
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA; Dept of Cell, Developmental and Cancer Biology, Oregon Health & Science University, USA
| | | | - Ugur Sezerman
- Eternans Ltd., UK; School of Medicine, Acibadem Mehmet Ali Aydinlar University, Atasehir Istanbul 34752, Turkey
| | - Gunseli Bayram Akcapinar
- Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Atasehir Istanbul 34752, Turkey; Eternans Ltd., UK
| | - Joshua C Saldivar
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA; Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, USA
| | - Emel Timucin
- Eternans Ltd., UK; School of Medicine, Acibadem Mehmet Ali Aydinlar University, Atasehir Istanbul 34752, Turkey
| | - Jared M Fischer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, USA; Dept of Molecular and Medical Genetics, Oregon Health & Science University, USA.
| |
Collapse
|
37
|
Zhang L, Pitcher LE, Prahalad V, Niedernhofer LJ, Robbins PD. Recent advances in the discovery of senolytics. Mech Ageing Dev 2021; 200:111587. [PMID: 34656616 DOI: 10.1016/j.mad.2021.111587] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/01/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022]
Abstract
The demonstration in model organisms that cellular senescence drives aging and age-related diseases has led to widespread efforts to identify compounds able to selectively kill senescent cells, termed senolytics. Approaches used to identify senolytics include bioinformatic analysis of senescent cell anti-apoptotic pathways (SCAPs) for drug development and screening of drugs libraries on different senescent cell types in culture. Alternatively, cytotoxic compounds can be made specific to senescent cells through a prodrug strategy such as linking the compound to a galactose moiety where toxicity is activated by lysosomal β-galactosidase. Identified senolytics can then be optimized through medicinal chemistry or linking to E3 targeting moieties to facilitate proteolysis of their targets. This review will provide an overview of approaches to identify senolytics and an update of the classes of senolytics identified to date.
Collapse
Affiliation(s)
- Lei Zhang
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Louise E Pitcher
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Vaishali Prahalad
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
38
|
Complex Positive Effects of SGLT-2 Inhibitor Empagliflozin in the Liver, Kidney and Adipose Tissue of Hereditary Hypertriglyceridemic Rats: Possible Contribution of Attenuation of Cell Senescence and Oxidative Stress. Int J Mol Sci 2021; 22:ijms221910606. [PMID: 34638943 PMCID: PMC8508693 DOI: 10.3390/ijms221910606] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: empagliflozin, sodium-glucose co-transporter 2 (SGLT-2) inhibitor, is an effective antidiabetic agent with strong cardio- and nephroprotective properties. The mechanisms behind its cardio- and nephroprotection are still not fully clarified. (2) Methods: we used male hereditary hypertriglyceridemic (hHTG) rats, a non-obese model of dyslipidaemia, insulin resistance, and endothelial dysfunction fed standard diet with or without empagliflozin for six weeks to explore the molecular mechanisms of empagliflozin effects. Nuclear magnetic resonance (NMR)-based metabolomics; quantitative PCR of relevant genes involved in lipid and glucose metabolism, or senescence; glucose and palmitic acid oxidation in isolated tissues and cell lines of adipocytes and hepatocytes were used. (3) Results: empagliflozin inhibited weight gain and decreased adipose tissue weight, fasting blood glucose, and triglycerides and increased HDL-cholesterol. It also improved insulin sensitivity in white fat. NMR spectroscopy identified higher plasma concentrations of ketone bodies, ketogenic amino acid leucine and decreased levels of pyruvate and alanine. In the liver, adipose tissue and kidney, empagliflozin up-regulated expression of genes involved in gluconeogenesis and down-regulated expression of genes involved in lipogenesis along with reduction of markers of inflammation, oxidative stress and cell senescence. (4) Conclusion: multiple positive effects of empagliflozin, including reduced cell senescence and oxidative stress, could contribute to its long-term cardio- and nephroprotective actions.
Collapse
|
39
|
Fialova JL, Raudenska M, Jakubek M, Kejik Z, Martasek P, Babula P, Matkowski A, Filipensky P, Masarik M. Novel Mitochondria-targeted Drugs for Cancer Therapy. Mini Rev Med Chem 2021; 21:816-832. [PMID: 33213355 DOI: 10.2174/1389557520666201118153242] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 11/22/2022]
Abstract
The search for mitochondria-targeted drugs has dramatically risen over the last decade. Mitochondria are essential organelles serving not only as a powerhouse of the cell but also as a key player in cell proliferation and cell death. Their central role in the energetic metabolism, calcium homeostasis and apoptosis makes them an intriguing field of interest for cancer pharmacology. In cancer cells, many mitochondrial signaling and metabolic pathways are altered. These changes contribute to cancer development and progression. Due to changes in mitochondrial metabolism and changes in membrane potential, cancer cells are more susceptible to mitochondria-targeted therapy. The loss of functional mitochondria leads to the arrest of cancer progression and/or a cancer cell death. Identification of mitochondrial changes specific for tumor growth and progression, rational development of new mitochondria-targeted drugs and research on delivery agents led to the advance of this promising area. This review will highlight the current findings in mitochondrial biology, which are important for cancer initiation, progression and resistance, and discuss approaches of cancer pharmacology with a special focus on the anti-cancer drugs referred to as 'mitocans'.
Collapse
Affiliation(s)
- Jindriska Leischner Fialova
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Martina Raudenska
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Milan Jakubek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Zdenek Kejik
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Pavel Martasek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, CZ-121 08 Prague, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| | - Adam Matkowski
- Department of Pharmaceutical Biology and Botany, Wroclaw Medical University, 50556 Borowska 211, Poland
| | - Petr Filipensky
- Department of Urology, St. Anne's Faculty Hospital, CZ-65691 Brno, Czech Republic
| | - Michal Masarik
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00 Brno, Czech Republic
| |
Collapse
|
40
|
Lagoumtzi SM, Chondrogianni N. Senolytics and senomorphics: Natural and synthetic therapeutics in the treatment of aging and chronic diseases. Free Radic Biol Med 2021; 171:169-190. [PMID: 33989756 DOI: 10.1016/j.freeradbiomed.2021.05.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/12/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022]
Abstract
Cellular senescence is a heterogeneous process guided by genetic, epigenetic and environmental factors, characterizing many types of somatic cells. It has been suggested as an aging hallmark that is believed to contribute to aging and chronic diseases. Senescent cells (SC) exhibit a specific senescence-associated secretory phenotype (SASP), mainly characterized by the production of proinflammatory and matrix-degrading molecules. When SC accumulate, a chronic, systemic, low-grade inflammation, known as inflammaging, is induced. In turn, this chronic immune system activation results in reduced SC clearance thus establishing a vicious circle that fuels inflammaging. SC accumulation represents a causal factor for various age-related pathologies. Targeting of several aging hallmarks has been suggested as a strategy to ameliorate healthspan and possibly lifespan. Consequently, SC and SASP are viewed as potential therapeutic targets either through the selective killing of SC or the selective SASP blockage, through natural or synthetic compounds. These compounds are members of a family of agents called senotherapeutics divided into senolytics and senomorphics. Few of them are already in clinical trials, possibly representing a future treatment of age-related pathologies including diseases such as atherosclerosis, osteoarthritis, osteoporosis, cancer, diabetes, neurodegenerative diseases such as Alzheimer's disease, cardiovascular diseases, hepatic steatosis, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis and age-related macular degeneration. In this review, we present the already identified senolytics and senomorphics focusing on their redox-sensitive properties. We describe the studies that revealed their effects on cellular senescence and enabled their nomination as novel anti-aging agents. We refer to the senolytics that are already in clinical trials and we present various adverse effects exhibited by senotherapeutics so far. Finally, we discuss aspects of the senotherapeutics that need improvement and we suggest the design of future senotherapeutics to target specific redox-regulated signaling pathways implicated either in the regulation of SASP or in the elimination of SC.
Collapse
Affiliation(s)
- Sofia M Lagoumtzi
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 116 35, Athens, Greece; Department of Biomedical Sciences, University of Western Attica, 28 Ag. Spyridonos Str., Egaleo, 12243, Athens, Greece.
| | - Niki Chondrogianni
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 116 35, Athens, Greece.
| |
Collapse
|
41
|
Ahumada-Castro U, Puebla-Huerta A, Cuevas-Espinoza V, Lovy A, Cardenas JC. Keeping zombies alive: The ER-mitochondria Ca 2+ transfer in cellular senescence. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119099. [PMID: 34274397 DOI: 10.1016/j.bbamcr.2021.119099] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 06/18/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence generates a permanent cell cycle arrest, characterized by apoptosis resistance and a pro-inflammatory senescence-associated secretory phenotype (SASP). Physiologically, senescent cells promote tissue remodeling during development and after injury. However, when accumulated over a certain threshold as happens during aging or after cellular stress, senescent cells contribute to the functional decline of tissues, participating in the generation of several diseases. Cellular senescence is accompanied by increased mitochondrial metabolism. How mitochondrial function is regulated and what role it plays in senescent cell homeostasis is poorly understood. Mitochondria are functionally and physically coupled to the endoplasmic reticulum (ER), the major calcium (Ca2+) storage organelle in mammalian cells, through special domains known as mitochondria-ER contacts (MERCs). In this domain, the release of Ca2+ from the ER is mainly regulated by inositol 1,4,5-trisphosphate receptors (IP3Rs), a family of three Ca2+ release channels activated by a ligand (IP3). IP3R-mediated Ca2+ release is transferred to mitochondria through the mitochondrial Ca2+ uniporter (MCU), where it modulates the activity of several enzymes and transporters impacting its bioenergetic and biosynthetic function. Here, we review the possible connection between ER to mitochondria Ca2+ transfer and senescence. Understanding the pathways that contribute to senescence is essential to reveal new therapeutic targets that allow either delaying senescent cell accumulation or reduce senescent cell burden to alleviate multiple diseases.
Collapse
Affiliation(s)
- Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Andrea Puebla-Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Victor Cuevas-Espinoza
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Alenka Lovy
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Department of Neuroscience, Center for Neuroscience Research, Tufts School of Medicine, Boston, MA, USA
| | - J Cesar Cardenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
42
|
Zhang JW, Zhang D, Yu BP. Senescent cells in cancer therapy: why and how to remove them. Cancer Lett 2021; 520:68-79. [PMID: 34237406 DOI: 10.1016/j.canlet.2021.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a stress response that imposes a growth arrest on cancer and nonmalignant cells during cancer therapy. By secreting a plethora of proinflammatory factors collectively termed the senescence-associated secretory phenotype (SASP), therapy-induced senescent cells can promote tumorigenesis. Moreover, the SASP from senescent cells is also able to drive therapy resistance and mediate many adverse effects of cancer therapy. Because senescent cell production often occurs during cancer therapy, it is important to carefully consider these potential detrimental effects. Senotherapy, which refers to selective removal of senescent cells, has been proposed as a promising adjuvant approach to eliminate the adverse effects of senescent cells. Thus, in this review we summarize in detail the mechanisms by which senescent cells contribute to tumorigenesis and therapeutic resistance. Also, we thoroughly discuss the potential strategies regarding how to effectively circumvent the undesirable effects of therapy-induced senescent cells.
Collapse
Affiliation(s)
- Jian-Wei Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, Hubei Province, People's Republic of China
| | - Dan Zhang
- Chongqing University Cancer Hospital, Chongqing, People's Republic of China
| | - Bao-Ping Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China; Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, Hubei Province, People's Republic of China.
| |
Collapse
|
43
|
Greenwood EK, Brown DR. Senescent Microglia: The Key to the Ageing Brain? Int J Mol Sci 2021; 22:4402. [PMID: 33922383 PMCID: PMC8122783 DOI: 10.3390/ijms22094402] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Ageing represents the single biggest risk factor for development of neurodegenerative disease. Despite being such long-lived cells, microglia have been relatively understudied for their role in the ageing process. Reliably identifying aged microglia has proven challenging, not least due to the diversity of cell populations, and the limitations of available models, further complicated by differences between human and rodent cells. Consequently, the literature contains multiple descriptions and categorisations of microglia with neurotoxic phenotypes, including senescence, without any unifying markers. The role of microglia in brain homeostasis, particularly iron storage and metabolism, may provide a key to reliable identification.
Collapse
Affiliation(s)
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK;
| |
Collapse
|
44
|
SMAD4 loss limits the vulnerability of pancreatic cancer cells to complex I inhibition via promotion of mitophagy. Oncogene 2021; 40:2539-2552. [PMID: 33686239 DOI: 10.1038/s41388-021-01726-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023]
Abstract
Pancreatic cancer is one of the deadliest forms of cancer, which is attributed to lack of effective treatment options and drug resistance. Mitochondrial inhibitors have emerged as a promising class of anticancer drugs, and several inhibitors of the electron transport chain (ETC) are being clinically evaluated. We hypothesized that resistance to ETC inhibitors from the biguanide class could be induced by inactivation of SMAD4, an important tumor suppressor involved in transforming growth factor β (TGFβ) signaling, and associated with altered mitochondrial activity. Here we show that, paradoxically, both TGFβ-treatment and the loss of SMAD4, a downstream member of TGFβ signaling cascade, induce resistance to biguanides, decrease mitochondrial respiration, and fragment the mitochondrial network. Mechanistically, the resistance of SMAD4-deficient cells is mediated by increased mitophagic flux driven by MAPK/ERK signaling, whereas TGFβ-induced resistance is autophagy-independent and linked to epithelial-to-mesenchymal transition (EMT). Interestingly, mitochondria-targeted tamoxifen, a complex I inhibitor under clinical trial, overcomes resistance mediated by SMAD4-deficiency or TGFβ signaling. Our data point to differential mechanisms underlying the resistance to treatment in PDAC arising from TGFβ signaling and SMAD4 loss, respectively. The findings will help the development of mitochondria-targeted therapy for pancreatic cancer patients with SMAD4 as a plausible predictive marker.
Collapse
|
45
|
Phadwal K, Vrahnas C, Ganley IG, MacRae VE. Mitochondrial Dysfunction: Cause or Consequence of Vascular Calcification? Front Cell Dev Biol 2021; 9:611922. [PMID: 33816463 PMCID: PMC8010668 DOI: 10.3389/fcell.2021.611922] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/04/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are crucial bioenergetics powerhouses and biosynthetic hubs within cells, which can generate and sequester toxic reactive oxygen species (ROS) in response to oxidative stress. Oxidative stress-stimulated ROS production results in ATP depletion and the opening of mitochondrial permeability transition pores, leading to mitochondria dysfunction and cellular apoptosis. Mitochondrial loss of function is also a key driver in the acquisition of a senescence-associated secretory phenotype that drives senescent cells into a pro-inflammatory state. Maintaining mitochondrial homeostasis is crucial for retaining the contractile phenotype of the vascular smooth muscle cells (VSMCs), the most prominent cells of the vasculature. Loss of this contractile phenotype is associated with the loss of mitochondrial function and a metabolic shift to glycolysis. Emerging evidence suggests that mitochondrial dysfunction may play a direct role in vascular calcification and the underlying pathologies including (1) impairment of mitochondrial function by mineral dysregulation i.e., calcium and phosphate overload in patients with end-stage renal disease and (2) presence of increased ROS in patients with calcific aortic valve disease, atherosclerosis, type-II diabetes and chronic kidney disease. In this review, we discuss the cause and consequence of mitochondrial dysfunction in vascular calcification and underlying pathologies; the role of autophagy and mitophagy pathways in preventing mitochondrial dysfunction during vascular calcification and finally we discuss mitochondrial ROS, DRP1, and HIF-1 as potential novel markers and therapeutic targets for maintaining mitochondrial homeostasis in vascular calcification.
Collapse
Affiliation(s)
- Kanchan Phadwal
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| | - Christina Vrahnas
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Ian G. Ganley
- Medical Research Council (MRC) Protein Phosphorylation and Ubiquitylation Unit, Sir James Black Centre, University of Dundee, Dundee, United Kingdom
| | - Vicky E. MacRae
- Functional Genetics and Development Division, The Roslin Institute and The Royal (Dick) School of Veterinary Studies (R(D)SVS), University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
46
|
Prašnikar E, Borišek J, Perdih A. Senescent cells as promising targets to tackle age-related diseases. Ageing Res Rev 2021; 66:101251. [PMID: 33385543 DOI: 10.1016/j.arr.2020.101251] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022]
Abstract
As the world's population progressively ages, the burden on the socio-economic and health systems is escalating, demanding sustainable and lasting solutions. Cellular senescence, one of the hallmarks of ageing, is a state of irreversible cell cycle arrest that occurs in response to various genotoxic stressors and is considered an important factor in the development of many age-related diseases and therefore a potential therapeutic target. Here, the role of senescent cells in age-related diseases is discussed, focusing on their formation and main characteristics. The mechanisms leading to senescent cells are presented, including replicative and premature senescence as well as senescence that occurs in various physiological processes, such as wound healing. The second part comprises a comprehensive description of various biomarkers currently used for the detection of senescent cells along with the investigated therapeutic approaches, namely senolytics, senomorphics and the clearance of senescent cells by the immune system. Potential delivery systems suitable for such therapies and model organisms to study senescence are also briefly examined. This in-depth overview of cellular senescence contributes to a deeper understanding of a rapidly evolving area aimed to tackle the age-related diseases in a more mechanistic way, as well as highlights future research opportunities.
Collapse
|
47
|
Bai Y, Yang J, Cui Y, Yao Y, Wu F, Liu C, Fan X, Zhang Y. Research Progress of Sirtuin4 in Cancer. Front Oncol 2021; 10:562950. [PMID: 33585187 PMCID: PMC7874138 DOI: 10.3389/fonc.2020.562950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/03/2020] [Indexed: 12/29/2022] Open
Abstract
Sirtuins (SIRTs) are members of the silent information regulator-2 family. They are a conserved family of nicotinamide adenine dinucleotide-dependent protein lysine deacylases. SIRTS are involved in intricate cellular processes. There are seven subtypes of SIRTs (1–7) in mammals. SIRT4 is located mainly in mitochondria and has various catalytic activities. These enzyme activities give it a diverse range of important biologic functions, such as energy metabolism, oxidative stress, and aging. Cancer is characterized as reprogramming of energy metabolism and redox imbalance, and SIRT4 can affect tumorigenesis. Here, we review the structure, localization, and enzyme activity of SIRT4 and its role in various neoplasms.
Collapse
Affiliation(s)
- Yibing Bai
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Jiani Yang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Ying Cui
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuanfei Yao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Feng Wu
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Caiqi Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaona Fan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
48
|
González-Gualda E, Baker AG, Fruk L, Muñoz-Espín D. A guide to assessing cellular senescence in vitro and in vivo. FEBS J 2021; 288:56-80. [PMID: 32961620 DOI: 10.1111/febs.15570] [Citation(s) in RCA: 300] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022]
Abstract
Cellular senescence is a physiological mechanism whereby a proliferating cell undergoes a stable cell cycle arrest upon damage or stress and elicits a secretory phenotype. This highly dynamic and regulated cellular state plays beneficial roles in physiology, such as during embryonic development and wound healing, but it can also result in antagonistic effects in age-related pathologies, degenerative disorders, ageing and cancer. In an effort to better identify this complex state, and given that a universal marker has yet to be identified, a general set of hallmarks describing senescence has been established. However, as the senescent programme becomes more defined, further complexities, including phenotype heterogeneity, have emerged. This significantly complicates the recognition and evaluation of cellular senescence, especially within complex tissues and living organisms. To address these challenges, substantial efforts are currently being made towards the discovery of novel and more specific biomarkers, optimized combinatorial strategies and the development of emerging detection techniques. Here, we compile such advances and present a multifactorial guide to identify and assess cellular senescence in cell cultures, tissues and living organisms. The reliable assessment and identification of senescence is not only crucial for better understanding its underlying biology, but also imperative for the development of diagnostic and therapeutic strategies aimed at targeting senescence in the clinic.
Collapse
Affiliation(s)
- Estela González-Gualda
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| | - Andrew G Baker
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Daniel Muñoz-Espín
- CRUK Cambridge Centre Early Detection Programme, Department of Oncology, Hutchison/MRC Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
49
|
Chatterjee S, Chakrabarty Y, Banerjee S, Ghosh S, Bhattacharyya SN. Mitochondria control mTORC1 activity-linked compartmentalization of eIF4E to regulate extracellular export of microRNAs. J Cell Sci 2020; 133:jcs250241. [PMID: 33262313 DOI: 10.1242/jcs.250241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
Defective intracellular trafficking and export of microRNAs (miRNAs) have been observed in growth-retarded mammalian cells having impaired mitochondrial potential and dynamics. Here, we found that uncoupling protein 2 (Ucp2)-mediated depolarization of mitochondrial membrane also results in progressive sequestration of miRNAs within polysomes and lowers their release via extracellular vesicles. Interestingly, the impaired miRNA-trafficking process in growth-retarded human cells could be reversed in the presence of Genipin, an inhibitor of Ucp2. Mitochondrial detethering of endoplasmic reticulum (ER), observed in cells with depolarized mitochondria, was found to be responsible for defective compartmentalization of translation initiation factor eIF4E to polysomes attached to ER. This caused a retarded translation process accompanied by enhanced retention of miRNAs and target mRNAs within ER-attached polysomes to restrict extracellular export of miRNAs. Reduced compartment-specific activity of the mammalian target of rapamycin complex 1 (mTORC1), the master regulator of protein synthesis, in cells with defective mitochondria or detethered ER, caused reduced phosphorylation of eIF4E-BP1 and prevented eIF4E targeting to ER-attached polysomes and miRNA export. These data suggest how mitochondrial membrane potential and dynamics, by affecting mTORC1 activity and compartmentalization, determine the subcellular localization and export of miRNAs.
Collapse
Affiliation(s)
- Susanta Chatterjee
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Yogaditya Chakrabarty
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Saikat Banerjee
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Souvik Ghosh
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
50
|
Lu Z, Zhang W, No YJ, Lu Y, Mirkhalaf Valashani SM, Rollet P, Jiang L, Ramaswamy Y, Dunstan CR, Jiang X, Zreiqat H. Baghdadite Ceramics Prevent Senescence in Human Osteoblasts and Promote Bone Regeneration in Aged Rats. ACS Biomater Sci Eng 2020; 6:6874-6885. [PMID: 33320606 DOI: 10.1021/acsbiomaterials.0c01120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Bone fractures and critical-sized bone defects present significant health threats for the elderly who have limited capacity for regeneration due to the presence of functionally compromised senescent cells. A wide range of synthetic materials has been developed to promote the regeneration of critical-sized bone defects, but it is largely unknown if a synthetic biomaterial (scaffold) can modulate cellular senescence and improve bone regeneration in aged scenarios. The current study investigates the interaction of Baghdadite (Ca3ZrSi2O9) ceramic scaffolds with senescent human primary osteoblast-like cells (HOBs) and its bone regeneration capacity in aged rats. A senescent HOB model was established by repeatedly passaging HOBs till passage 7 (P7). Compared to the clinically used hydroxyapatite/tricalcium phosphate (HA/TCP), Baghdadite prevented senescence induction in P7 HOBs and markedly negated the paracrine effect of P7 HOB secretomes that mediated the up-regulations of cellular senescence-associated gene expression levels in P2 HOBs. We further demonstrated that conditioned media extracted from Baghdadite corrected the dysfunctional mitochondria in P7 HOBs. In vivo, the bone regeneration capacity was enhanced when 3D printed Baghdadite scaffolds were implanted in a calvaria critical-sized bone defect model in both young and aged rats compared to HA/TCP scaffolds, but a better effect was observed in aged rats than in young rats. This study suggests that Baghdadite ceramic represents a novel and promising biomaterial approach to promote bone regeneration capacity in the elderly by providing an anti-senescent microenvironment.
Collapse
Affiliation(s)
- ZuFu Lu
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - WenJie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,National Clinical Research Center of Stomatology, Shanghai 200011, China.,Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Young Jung No
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Yuezhi Lu
- Department of Prosthodontics, Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,National Clinical Research Center of Stomatology, Shanghai 200011, China.,Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Seyed Mohammad Mirkhalaf Valashani
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Paul Rollet
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Liting Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,National Clinical Research Center of Stomatology, Shanghai 200011, China.,Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Yogambha Ramaswamy
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Colin R Dunstan
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - XinQuan Jiang
- Department of Prosthodontics, Shanghai Ninth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.,National Clinical Research Center of Stomatology, Shanghai 200011, China.,Oral Bioengineering and Regenerative Medicine Lab, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Hala Zreiqat
- Tissue Engineering & Biomaterials Research Unit, School of Biomedical Engineering, The University of Sydney, Sydney, NSW 2006, Australia.,ARC Training Centre for Innovative BioEngineering, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|