1
|
Ding W, Bao S, Zhao Q, Hao W, Fang K, Xiao Y, Lin X, Zhao Z, Xu X, Cui X, Yang X, Yao L, Jin H, Zhang K, Guo J. Blocking ACSL6 Compromises Autophagy via FLI1-Mediated Downregulation of COLs to Radiosensitize Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403202. [PMID: 39206814 PMCID: PMC11516120 DOI: 10.1002/advs.202403202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer (LC) is the leading cause of cancer-related mortality worldwide. Radiotherapy is the main component of LC treatment; however, its efficacy is often limited by radioresistance development, resulting in unsatisfactory clinical outcomes. Here, we found that LC radiosensitivity is up-regulated by decreased expression of long-chain acyl-CoA synthase 6 (ACSL6) after irradiation. Deletion of ACSL6 results in significant elevation of Friend leukemia integration 1 transcription factor (FLI1) and a marked decline of collagens (COLs). Blocking of ACSL6 impairs the tumor growth and upregulates FLI1, which reduces the levels of COLs and compromises irradiation-induced autophagy, leading to considerable therapeutic benefits during radiotherapy. Moreover, the direct interaction between ACSL6 and FLI1 and engagement between FLI1 and COLs indicates the involvement of the ACSL6-FLI1-COL axis. Finally, the potently adjusted autophagy flux reduces its otherwise contributive capability in surviving irradiation stress and leads to satisfactory radiosensitization for LC radiotherapy. These results demonstrate that enhanced ACSL6 expression promotes the aggressive performance of irradiated LC through increased FLI1-COL-mediated autophagy flux. Thus, the ACSL6-FLI1-Col-autophagy axis may be targeted to enhance the radiosensitivity of LC and improve the management of LC in radiotherapy.
Collapse
Affiliation(s)
- Wen Ding
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Shijun Bao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Qingwei Zhao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Wei Hao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Kai Fang
- Department of Medicine CollegeJiangnan UniversityWuxiJiangsu214000P. R. China
| | - Yanlan Xiao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xiaoting Lin
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Zhemeng Zhao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinyi Xu
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- College of Basic MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinyue Cui
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xiwen Yang
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Liuhuan Yao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Hai Jin
- Department of Cardiothoracic SurgeryChanghai HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Kun Zhang
- Department of Laboratory Medicine and Central LaboratorySichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072P. R. China
| | - Jiaming Guo
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| |
Collapse
|
2
|
Bouley SJ, Grassetti AV, Allaway RJ, Wood MD, Hou HW, Burdon Dasbach IR, Seibel W, Wu J, Gerber SA, Dragnev KH, Walker JA, Sanchez Y. Chemical genetic screens reveal defective lysosomal trafficking as synthetic lethal with NF1 loss. J Cell Sci 2024; 137:jcs262343. [PMID: 39016685 PMCID: PMC11361638 DOI: 10.1242/jcs.262343] [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: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Neurofibromatosis type 1, a genetic disorder caused by pathogenic germline variations in NF1, predisposes individuals to the development of tumors, including cutaneous and plexiform neurofibromas (CNs and PNs), optic gliomas, astrocytomas, juvenile myelomonocytic leukemia, high-grade gliomas and malignant peripheral nerve sheath tumors (MPNSTs), which are chemotherapy- and radiation-resistant sarcomas with poor survival. Loss of NF1 also occurs in sporadic tumors, such as glioblastoma (GBM), melanoma, breast, ovarian and lung cancers. We performed a high-throughput screen for compounds that were synthetic lethal with NF1 loss, which identified several leads, including the small molecule Y102. Treatment of cells with Y102 perturbed autophagy, mitophagy and lysosome positioning in NF1-deficient cells. A dual proteomics approach identified BLOC-one-related complex (BORC), which is required for lysosome positioning and trafficking, as a potential target of Y102. Knockdown of a BORC subunit using siRNA recapitulated the phenotypes observed with Y102 treatment. Our findings demonstrate that BORC might be a promising therapeutic target for NF1-deficient tumors.
Collapse
Affiliation(s)
- Stephanie J. Bouley
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Andrew V. Grassetti
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Department of Biochemistry and Cellular Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Robert J. Allaway
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Matthew D. Wood
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Helen W. Hou
- Department of Pharmacology and Toxicology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - India R. Burdon Dasbach
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - William Seibel
- Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jimmy Wu
- Department of Chemistry, Dartmouth College, Hanover, NH 03755, USA
| | - Scott A. Gerber
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Department of Biochemistry and Cellular Biology, Geisel School of Medicine, Hanover, NH 03755, USA
| | - Konstantin H. Dragnev
- Department of Medicine, Geisel School of Medicine, Hanover, NH 03755, USA
- Section of Medical Oncology, Geisel School of Medicine, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766, USA
| | - James A. Walker
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yolanda Sanchez
- Department of Molecular and Systems Biology, Geisel School of Medicine, Hanover, NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03766, USA
| |
Collapse
|
3
|
Liu Y, Lomeli I, Kron SJ. Therapy-Induced Cellular Senescence: Potentiating Tumor Elimination or Driving Cancer Resistance and Recurrence? Cells 2024; 13:1281. [PMID: 39120312 PMCID: PMC11312217 DOI: 10.3390/cells13151281] [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: 06/01/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Cellular senescence has been increasingly recognized as a hallmark of cancer, reflecting its association with aging and inflammation, its role as a response to deregulated proliferation and oncogenic stress, and its induction by cancer therapies. While therapy-induced senescence (TIS) has been linked to resistance, recurrence, metastasis, and normal tissue toxicity, TIS also has the potential to enhance therapy response and stimulate anti-tumor immunity. In this review, we examine the Jekyll and Hyde nature of senescent cells (SnCs), focusing on how their persistence while expressing the senescence-associated secretory phenotype (SASP) modulates the tumor microenvironment through autocrine and paracrine mechanisms. Through the SASP, SnCs can mediate both resistance and response to cancer therapies. To fulfill the unmet potential of cancer immunotherapy, we consider how SnCs may influence tumor inflammation and serve as an antigen source to potentiate anti-tumor immune response. This new perspective suggests treatment approaches based on TIS to enhance immune checkpoint blockade. Finally, we describe strategies for mitigating the detrimental effects of senescence, such as modulating the SASP or targeting SnC persistence, which may enhance the overall benefits of cancer treatment.
Collapse
Affiliation(s)
| | | | - Stephen J. Kron
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Zhuang X, Ruan J, Zhou C, Li Z. The emerging and diverse roles of F-box proteins in spermatogenesis and male infertility. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:13. [PMID: 38918264 PMCID: PMC11199460 DOI: 10.1186/s13619-024-00196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
F-box proteins play essential roles in various cellular processes of spermatogenesis by means of ubiquitylation and subsequent target protein degradation. They are the substrate-recognition subunits of SKP1-cullin 1-F-box protein (SCF) E3 ligase complexes. Dysregulation of F‑box protein‑mediated proteolysis could lead to male infertility in humans and mice. The emerging studies revealed the physiological function, pathological evidence, and biochemical substrates of F-box proteins in the development of male germ cells, which urging us to review the current understanding of how F‑box proteins contribute to spermatogenesis. More functional and mechanistic study will be helpful to define the roles of F-box protein in spermatogenesis, which will pave the way for the logical design of F-box protein-targeted diagnosis and therapies for male infertility, as the spermatogenic role of many F-box proteins remains elusive.
Collapse
Affiliation(s)
- Xuan Zhuang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, 363000, China
- Department of Urology, the First Affiliated Hospital of Xiamen University, Xiamen, Fujian, 361003, China
| | - Jun Ruan
- College of Life Sciences, Central China Normal University, Wuhan, Hubei, 430079, China
| | - Canquan Zhou
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangdong Provincial Clinical Research Center for obstetrical and gynecological diseases, Center for Reproductive Medicine and Department of Gynecology & Obstetrics, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhiming Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
5
|
Chouhan NK, Eedara A, Talati MN, Ambadipudi SSSSS, Andugulapati SB, Pabbaraja S. Glucosyltriazole amphiphile treatment attenuates breast cancer by modulating the AMPK signaling. Drug Dev Res 2024; 85:e22215. [PMID: 38837718 DOI: 10.1002/ddr.22215] [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: 02/12/2024] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 06/07/2024]
Abstract
Breast cancer is the second most frequent cancer among women. Out of various subtypes, triple-negative breast cancers (TNBCs) account for 15% of breast cancers and exhibit more aggressive characteristics as well as a worse prognosis due to their proclivity for metastatic progression and limited therapeutic strategies. It has been demonstrated that AMP-activated protein kinase (AMPK) has context-specific protumorigenic implications in breast cancer cells. A set of glucosyltriazole amphiphiles, consisting of acetylated (9a-h) and unmodified sugar hydroxyl groups (10a-h), were synthesized and subjected to in vitro biological evaluation. Among them, 9h exhibited significant anticancer activity against MDA-MB-231, MCF-7, and 4T1 cell lines with IC50 values of 12.5, 15, and 12.55 μM, respectively. Further, compound 9h was evaluated for apoptosis and cell cycle analysis in in vitro models (using breast cancer cells) and antitumour activity in an in vivo model (orthotopic mouse model using 4T1 cells). Annexin-V assay results revealed that treatment with 9h caused 34% and 28% cell death at a concentration of 15 or 7.5 μM, respectively, while cell cycle analysis demonstrated that 9h arrested the cells at the G2/M or G1 phase in MCF-7, MDA-MB-231 and 4T1 cells, respectively. Further, in vivo, investigation showed that compound 9h exhibited equipotent as doxorubicin at 7.5 mg/kg, and superior efficacy than doxorubicin at 15 mg/kg. The mechanistic approach revealed that 9h showed potent anticancer activity in an in vivo orthotopic model (4T1 cells) partly by suppressing the AMPK activation. Therefore, modulating the AMPK activation could be a probable approach for targeting breast cancer and mitigating cancer progression.
Collapse
Affiliation(s)
- Neeraj Kumar Chouhan
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Abhisheik Eedara
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Mamta N Talati
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sudha S S S S Ambadipudi
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Sai Balaji Andugulapati
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology (IICT), Hyderabad, India
| | - Srihari Pabbaraja
- Department of Organic Synthesis & Process Chemistry, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Zhang Q, Lou Y, Fang H, Sun S, Jin R, Ji Y, Chen Z. Cancer‑associated fibroblasts under therapy‑induced senescence in the tumor microenvironment (Review). Exp Ther Med 2024; 27:150. [PMID: 38476922 PMCID: PMC10928991 DOI: 10.3892/etm.2024.12438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
Current cancer treatments target tumor cells; however, the tumor microenvironment (TME) induces therapeutic resistance, tumor development and metastasis, thus rendering these treatments ineffective. Research on the TME has therefore concentrated on nonmalignant cells. Cancer-associated fibroblasts (CAFs) are a major TME component, which contribute to cancer progression due to their diverse origins, phenotypes and functions, including cancer cell invasion and migration, extracellular matrix remodeling, tumor metabolism modulation and therapeutic resistance. Standard cancer treatment typically exacerbates the senescence-associated secretory phenotype (SASP) of senescent cancer cells and nonmalignant cells that actively leak proinflammatory signals in the TME. Therapy-induced senescence may impair cancer cell activity and compromise treatment responsiveness. CAFs and SASP are well-studied in the formation and progression of cancer. The present review discusses the current data on CAF senescence caused by anticancer treatment and assesses how senescence-like CAFs affect tumor formation. The development of senolytic medication for aging stromal cells is also highlighted. Combining cancer therapies with senolytics may boost therapeutic effects and provide novel possibilities for research.
Collapse
Affiliation(s)
- Qiuhua Zhang
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yijie Lou
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Hao Fang
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Shaopeng Sun
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Rijuan Jin
- Department of Oncology, First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Yunxi Ji
- Department of General Practice, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310003, P.R. China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Institute of Cancer Research, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
7
|
Henrich LM, Greimelmaier K, Wessolly M, Klopp NA, Mairinger E, Krause Y, Berger S, Wohlschlaeger J, Schildhaus HU, Baba HA, Mairinger FD, Borchert S. The Impact of Cancer-Associated Fibroblasts on the Biology and Progression of Colorectal Carcinomas. Genes (Basel) 2024; 15:209. [PMID: 38397199 PMCID: PMC10888097 DOI: 10.3390/genes15020209] [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: 01/03/2024] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Colorectal cancer (CRC) is a leading cause of cancer-related deaths globally. Cancer-associated fibroblasts (CAFs) are major components of CRC's tumour microenvironment (TME), but their biological background and interplay with the TME remain poorly understood. This study investigates CAF biology and its impact on CRC progression. (2) The cohort comprises 155 cases, including CRC, with diverse localizations, adenomas, inflammations, and controls. Digital gene expression analysis examines genes associated with signalling pathways (MAPK, PI3K/Akt, TGF-β, WNT, p53), while next-generation sequencing (NGS) determines CRC mutational profiles. Immunohistochemical FAP scoring assesses CAF density and activity. (3) FAP expression is found in 81 of 150 samples, prevalent in CRC (98.4%), adenomas (27.5%), and inflammatory disease (38.9%). Several key genes show significant associations with FAP-positive fibroblasts. Gene set enrichment analysis (GSEA) highlights PI3K and MAPK pathway enrichment alongside the activation of immune response pathways like natural killer (NK)-cell-mediated cytotoxicity via CAFs. (4) The findings suggest an interplay between CAFs and cancer cells, influencing growth, invasiveness, angiogenesis, and immunogenicity. Notably, TGF-β, CDKs, and the Wnt pathway are affected. In conclusion, CAFs play a significant role in CRC and impact the TME throughout development.
Collapse
Affiliation(s)
- Larissa Maria Henrich
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Kristina Greimelmaier
- Department of Pathology, Diakonissenkrankenhaus Flensburg, 24939 Flensburg, Germany (J.W.)
| | - Michael Wessolly
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Nick Alexander Klopp
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Elena Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Yvonne Krause
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Sophia Berger
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Jeremias Wohlschlaeger
- Department of Pathology, Diakonissenkrankenhaus Flensburg, 24939 Flensburg, Germany (J.W.)
| | - Hans-Ulrich Schildhaus
- Targos-A Discovery Life Sciences Company, Germaniastraße 7, 34119 Kassel, Germany;
- Institute of Pathology Nordhessen, Germaniastraße 7, 34119 Kassel, Germany
| | - Hideo Andreas Baba
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Fabian Dominik Mairinger
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| | - Sabrina Borchert
- Institute of Pathology, University Hospital Essen, University of Duisburg Essen, 45147 Essen, Germany; (L.M.H.); (M.W.); (N.A.K.); (E.M.); (H.A.B.); (S.B.)
| |
Collapse
|
8
|
Krasnova O, Kovaleva A, Saveleva A, Kulakova K, Bystrova O, Martynova M, Domnina A, Sopova J, Neganova I. Mesenchymal stem cells lose the senescent phenotype under 3D cultivation. Stem Cell Res Ther 2023; 14:373. [PMID: 38111010 PMCID: PMC10729581 DOI: 10.1186/s13287-023-03599-8] [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: 08/03/2023] [Accepted: 12/04/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Three-dimensional (3D) cell culture is widely used in various fields of cell biology. In comparison to conventional two-dimensional (2D) cell culture, 3D cell culture facilitates a more accurate replication of the in vivo microenvironment, which is essential for obtaining more relevant results. The application of 3D cell culture techniques in regenerative medicine, particularly in mesenchymal stem cell (MSC)-based research, has been extensively studied. Many of these studies focus on the enhanced paracrine activity of MSCs cultured in 3D environments. However, few focus on the cellular processes that occur during 3D cultivation. METHODS In this work, we studied the changes occurring within 3D-cultured MSCs (3D-MSCs). Specifically, we examined the expression of numerous senescent-associated markers, the actin cytoskeleton structure, the architecture of the Golgi apparatus and the localization of mTOR, one of the main positive regulators of replicative senescence. In addition, we assessed whether the selective elimination of senescent cells occurs upon 3D culturing by using cell sorting based on autofluorescence. RESULTS Our findings indicate that 3D-MSCs were able to lose replicative senescence markers under 3D cell culture conditions. We observed changes in actin cytoskeleton structure, Golgi apparatus architecture and revealed that 3D cultivation leads to the nuclear localization of mTOR, resulting in a decrease in its active cytoplasmic form. Additionally, our findings provide evidence that 3D cell culture promotes the phenotypic reversion of senescent cell phenotype rather than their removal from the bulk population. CONCLUSION These novel insights into the biology of 3D-MSCs can be applied to research in regenerative medicine to overcome replicative senescence and MSC heterogeneity as they often pose significant concerns regarding safety and effectiveness for therapeutic purposes.
Collapse
Affiliation(s)
- O Krasnova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia.
| | - A Kovaleva
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - A Saveleva
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - K Kulakova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - O Bystrova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - M Martynova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - A Domnina
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - J Sopova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - I Neganova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| |
Collapse
|
9
|
Bulak K, Kycko A, Śmiech A, Łopuszyński W. Immunoreactivity of p21, MMP-1 and CB2 receptor proteins in cutaneous canine mast cell tumours: an association with the three-tier grading system. J Vet Res 2023; 67:611-618. [PMID: 38130452 PMCID: PMC10730558 DOI: 10.2478/jvetres-2023-0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Mast cell tumours (MCTs) arise in the dermis and subcutaneous tissues in animals and humans and are one of the most common neoplasms of the skin in dogs. Cannabinoid type 2 receptor (CB2R), cyclin-dependent kinase inhibitor (p21) and matrix metalloproteinase 1 (MMP-1) are potential targets for novel anti-tumour therapeutic strategies. This study evaluated by immunohistochemical means the reactivity of p21, MMP-1 and CB2R proteins in association with a three-tier grading system in cutaneous canine MCTs. Material and Methods Formalin-fixed, paraffin-embedded canine MCTs were processed for histochemical analysis and immunohistochemical staining using antibodies against p21, MMP-1 and CB2R. The results were analysed statistically. Results The strongest p21 immunolabelling was detected in grade 3 MCTs, while grade 1 tumours showed mild or no detectable p21 immunoreactivity (P-value < 0.001). Strong immunolabelling of MMP-1 was the most common in grade 1 tumours (P-value < 0.001) and CB2R was significantly less frequent in grade 3 tumours than in grade 1 (P-value < 0.001) and grade 2 (P-value < 0.001). Conclusion High immunoreactivity of MMP-1 can be a marker of grade 1 MCTs in dogs, whereas p21 protein overexpression can be a marker of grade 3 canine MCTs. Strong CB2R immunoreactivity with simultaneous underexpression of p21 and high immunoreactivity of MMP-1 proteins may indicate that the use of cannabinoids in grade 1 MCTs in dogs is practicable.
Collapse
Affiliation(s)
- Kamila Bulak
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences in Lublin, 20-612Lublin, Poland
| | - Anna Kycko
- Department of Pathology, National Veterinary Research Institute, 24-100Puławy, Poland
| | - Anna Śmiech
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences in Lublin, 20-612Lublin, Poland
| | - Wojciech Łopuszyński
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences in Lublin, 20-612Lublin, Poland
| |
Collapse
|
10
|
Hela F, Aguayo-Mazzucato C. Interaction between Autophagy and Senescence in Pancreatic Beta Cells. BIOLOGY 2023; 12:1205. [PMID: 37759604 PMCID: PMC10525299 DOI: 10.3390/biology12091205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023]
Abstract
Aging leads to an increase in cellular stress due to the fragility of the organism and the inability to cope with it. In this setting, there is a higher chance of developing different cardiometabolic diseases like diabetes. Cellular senescence and autophagy, both hallmarks of aging and stress-coping mechanisms, have gained increased attention for their role in the pathophysiology of diabetes. Studies show that impairing senescence dampens and even prevents diabetes while the role of autophagy is more contradictory, implying a context- and disease-stage-dependent effect. Reports show conflicting data about the effect of autophagy on senescence while the knowledge about this interaction in beta cells remains scarce. Elucidating this interaction between autophagy and senescence in pancreatic beta cells will lead to an identification of their respective roles and the extent of the effect each mechanism has on beta cells and open new horizons for developing novel therapeutic agents. To help illuminate this relationship we will review the latest findings of cellular senescence and autophagy with a special emphasis on pancreatic beta cells and diabetes.
Collapse
Affiliation(s)
| | - Cristina Aguayo-Mazzucato
- Section on Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
11
|
Zhou H, Wang J, Wen T. The molecular neural mechanism underlying the acceleration of brain aging due to Dcf1 deficiency. Mol Cell Neurosci 2023; 126:103884. [PMID: 37506857 DOI: 10.1016/j.mcn.2023.103884] [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: 05/01/2023] [Revised: 07/11/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023] Open
Abstract
Owing to the continuous increase in human life expectancy, the management of aging-related diseases has become an urgent issue. The brain dominates the central nervous system; therefore, brain aging is a key area of aging-related research. We previously uncovered that dendritic cell factor 1 (Dcf1) maintains the stemness of neural stem cells and its expression in Drosophila can prolong lifespan, suggesting an association between Dcf1 and aging; however, the specific underlying neural mechanism remains unclear. In the present study, we show for the first time that hippocampal neurogenesis is decreased in aged Dcf1-/- mice, which leads to a decrease in the number of brain neurons and an increased number of senescent cells. Moreover, astrocytes proliferate abnormally and express elevated mRNA levels of aging-related factors, in addition to displaying increased activation of Akt and Foxo3a. Finally, behavioral tests confirm that aged Dcf1-/- mice exhibit a significant decline in cognitive abilities related to learning and memory. In conclusion, we reveal a novel mechanism underlying brain aging triggered by Dcf1 deficiency at the molecular, cellular, tissue, and behavioral levels, providing a new perspective for the exploration of brain aging.
Collapse
Affiliation(s)
- Haicong Zhou
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, China; Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China
| | - Tieqiao Wen
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University Shanghai, China.
| |
Collapse
|
12
|
Luo X, Zhang R, Schefczyk S, Liang Y, Lin SS, Liu S, Baba HA, Lange CM, Wedemeyer H, Lu M, Broering R. Nuclear translocation of YAP drives BMI-associated hepatocarcinogenesis in hepatitis B virus infection. Liver Int 2023; 43:2002-2016. [PMID: 37312627 DOI: 10.1111/liv.15628] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIMS Hepatitis B virus (HBV) infection is a major cause of hepatocellular carcinoma (HCC) development and progression. The aim of this study was to mechanistically investigate the involvement of Hippo signalling in HBV surface antigen (HBsAg)-dependent neoplastic transformation. METHODS Liver tissue and hepatocytes from HBsAg-transgenic mice were examined for the Hippo cascade and proliferative events. Functional experiments in mouse hepatoma cells included knockdown, overexpression, luciferase reporter assays and chromatin immunoprecipitation. Results were validated in HBV-related HCC biopsies. RESULTS Hepatic expression signatures in HBsAg-transgenic mice correlated with YAP responses, cell cycle control, DNA damage and spindle events. Polyploidy and aneuploidy occurred in HBsAg-transgenic hepatocytes. Suppression and inactivation of MST1/2 led to the loss of YAP phosphorylation and the induction of BMI1 expression in vivo and in vitro. Increased BMI1 directly mediated cell proliferation associated with decreased level of p16INK4a , p19ARF , p53 and Caspase 3 as well as increased Cyclin D1 and γ-H2AX expression. Chromatin immunoprecipitation and the analysis of mutated binding sites in dual-luciferase reporter assays confirmed that the YAP/TEAD4 transcription factor complex bound and activated the Bmi1 promoter. In chronic hepatitis B patients, paired liver biopsies of non-tumour and tumour tissue indicated a correlation between YAP expression and the abundance of BMI1. In a proof-of-concept, treatment of HBsAg-transgenic mice with YAP inhibitor verteporfin directly suppressed the BMI1-related cell cycle. CONCLUSION HBV-associated proliferative HCC might be related to the HBsAg-YAP-BMI1 axis and offer a potential target for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Xufeng Luo
- Institute for Lymphoma Research, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Rui Zhang
- Department of Hepato-Pancreato-Biliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Stefan Schefczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Yaojie Liang
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Shu S Lin
- Department of Hepato-Pancreato-Biliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian M Lange
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Internal Medicine II, LMU University Hospital Munich, Munich, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
Wu J, Kramer K, Crowe DL. Lysine metabolism is a novel metabolic tumor suppressor pathway in breast cancer. Oncogene 2023:10.1038/s41388-023-02766-8. [PMID: 37393340 DOI: 10.1038/s41388-023-02766-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023]
Abstract
The International Agency for Research on Cancer determined that obesity is the primary preventable cause of breast cancer. The nuclear receptor peroxisome proliferator activated receptor γ (PPARγ) binds inflammatory mediators in obesity and its expression is reduced in human breast cancer. We created a new model to better understand how the obese microenvironment alters nuclear receptor function in breast cancer. The obesity related cancer phenotype was PPARγ dependent; deletion of PPARγ in mammary epithelium which is a tumor suppressor in lean mice unexpectedly increased tumor latency, reduced the luminal progenitor (LP) tumor cell fraction, and increased autophagic and senescent cells. Loss of PPARγ expression in mammary epithelium of obese mice increased expression of 2-aminoadipate semialdehyde synthase (AASS) which regulates lysine catabolism to acetoacetate. PPARγ-associated co-repressors and activators regulated AASS expression via a canonical response element. AASS expression was significantly reduced in human breast cancer, and AASS overexpression or acetoacetate treatment inhibited proliferation and induced autophagy and senescence in human breast cancer cell lines. Genetic or pharmacologic HDAC inhibition promoted autophagy and senescence in mammary tumor cells in vitro and in vivo. We concluded that lysine metabolism is a novel metabolic tumor suppressor pathway in breast cancer.
Collapse
Affiliation(s)
- Jianchun Wu
- University of Illinois Cancer Center, 801 S. Paulina Street, Room 525, Chicago, IL, 60612, USA
| | - Kaitrin Kramer
- University of Illinois Cancer Center, 801 S. Paulina Street, Room 525, Chicago, IL, 60612, USA
| | - David L Crowe
- University of Illinois Cancer Center, 801 S. Paulina Street, Room 525, Chicago, IL, 60612, USA.
| |
Collapse
|
14
|
Zhang J, Xiang Q, Wu M, Lao YZ, Xian YF, Xu HX, Lin ZX. Autophagy Regulators in Cancer. Int J Mol Sci 2023; 24:10944. [PMID: 37446120 PMCID: PMC10341480 DOI: 10.3390/ijms241310944] [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: 04/25/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Autophagy plays a complex impact role in tumor initiation and development. It serves as a double-edged sword by supporting cell survival in certain situations while also triggering autophagic cell death in specific cellular contexts. Understanding the intricate functions and mechanisms of autophagy in tumors is crucial for guiding clinical approaches to cancer treatment. Recent studies highlight its significance in various aspects of cancer biology. Autophagy enables cancer cells to adapt to and survive unfavorable conditions by recycling cellular components. However, excessive or prolonged autophagy can lead to the self-destruction of cancer cells via a process known as autophagic cell death. Unraveling the molecular mechanisms underlying autophagy regulation in cancer is crucial for the development of targeted therapeutic interventions. In this review, we seek to present a comprehensive summary of current knowledge regarding autophagy, its impact on cancer cell survival and death, and the molecular mechanisms involved in the modulation of autophagy for cancer therapy.
Collapse
Affiliation(s)
- Juan Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (J.Z.); (Y.-F.X.)
| | - Qian Xiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Q.X.); (M.W.); (Y.-Z.L.)
| | - Man Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Q.X.); (M.W.); (Y.-Z.L.)
| | - Yuan-Zhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Q.X.); (M.W.); (Y.-Z.L.)
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (J.Z.); (Y.-F.X.)
| | - Hong-Xi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Q.X.); (M.W.); (Y.-Z.L.)
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (J.Z.); (Y.-F.X.)
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
15
|
Cayo A, Venturini W, Rebolledo-Mira D, Moore-Carrasco R, Herrada AA, Nova-Lamperti E, Valenzuela C, Brown NE. Palbociclib-Induced Cellular Senescence Is Modulated by the mTOR Complex 1 and Autophagy. Int J Mol Sci 2023; 24:ijms24119284. [PMID: 37298236 DOI: 10.3390/ijms24119284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/17/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
Despite not dividing, senescent cells acquire the ability to synthesize and secrete a plethora of bioactive molecules, a feature known as the senescence-associated secretory phenotype (SASP). In addition, senescent cells often upregulate autophagy, a catalytic process that improves cell viability in stress-challenged cells. Notably, this "senescence-related autophagy" can provide free amino acids for the activation of mTORC1 and the synthesis of SASP components. However, little is known about the functional status of mTORC1 in models of senescence induced by CDK4/6 inhibitors (e.g., Palbociclib), or the effects that the inhibition of mTORC1 or the combined inhibition of mTORC1 and autophagy have on senescence and the SASP. Herein, we examined the effects of mTORC1 inhibition, with or without concomitant autophagy inhibition, on Palbociclib-driven senescent AGS and MCF-7 cells. We also assessed the pro-tumorigenic effects of conditioned media from Palbociclib-driven senescent cells with the inhibition of mTORC1, or with the combined inhibition of mTORC1 and autophagy. We found that Palbociclib-driven senescent cells display a partially reduced activity of mTORC1 accompanied by increased levels of autophagy. Interestingly, further mTORC1 inhibition exacerbated the senescent phenotype, a phenomenon that was reversed upon autophagy inhibition. Finally, the SASP varied upon inhibiting mTORC1, or upon the combined inhibition of mTORC1 and autophagy, generating diverse responses in cell proliferation, invasion, and migration of non-senescent tumorigenic cells. Overall, variations in the SASP of Palbociclib-driven senescent cells with the concomitant inhibition of mTORC1 seem to depend on autophagy.
Collapse
Affiliation(s)
- Angel Cayo
- Center for Medical Research, School of Medicine, University of Talca, Talca 3460000, Chile
- Institute for Interdisciplinary Research, Academic Vice Rectory, University of Talca, Talca 3460000, Chile
| | - Whitney Venturini
- Center for Medical Research, School of Medicine, University of Talca, Talca 3460000, Chile
- Institute for Interdisciplinary Research, Academic Vice Rectory, University of Talca, Talca 3460000, Chile
| | - Danitza Rebolledo-Mira
- Center for Medical Research, School of Medicine, University of Talca, Talca 3460000, Chile
| | - Rodrigo Moore-Carrasco
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, University of Talca, Talca 3460000, Chile
| | - Andrés A Herrada
- Lymphatic and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca 3467987, Chile
| | - Estefanía Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Department of Clinical Biochemistry and Immunology, Pharmacy Faculty, Universidad de Concepción, Concepción 4070386, Chile
| | - Claudio Valenzuela
- Center for Medical Research, School of Medicine, University of Talca, Talca 3460000, Chile
| | - Nelson E Brown
- Center for Medical Research, School of Medicine, University of Talca, Talca 3460000, Chile
| |
Collapse
|
16
|
Qin A. An anti-cancer surveillance by the interplay between interferon-beta and retinoblastoma protein RB1. Front Oncol 2023; 13:1173467. [PMID: 37182173 PMCID: PMC10174298 DOI: 10.3389/fonc.2023.1173467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 05/16/2023] Open
Abstract
Interferon-beta (IFN-β), an extracellular cytokine that initiates signaling pathways for gene regulation, has been demonstrated to function as a tumor suppressor protein through lentiviral gene transduction. In this article, I review the relevant previous works and propose a cell cycle-based, tumor suppressor protein-mediated mechanism of anti-cancer surveillance. IFN-β induces a tumor cell cycle alteration that leads to S phase accumulation, senescence entry, and a loss of tumorigenicity in solid tumor cells. IFN-β does not show a significant cell cycle effect in their normal counterparts. Retinoblastoma protein RB1, another tumor suppressor protein, tightly controls the cell cycle and differentiation of normal cells, preventing them from being significantly impacted by the IFN-β effect. The interplay between IFN-β and RB1 acts as a mechanism of cell cycle-based, tumor suppressor protein-mediated anti-cancer surveillance that can selectively suppress solid tumor or proliferating transformed cells from the loss of control leading to cancer. This mechanism has important implications for the treatment of solid tumors.
Collapse
Affiliation(s)
- Albert Qin
- Medical Research & Clinical Operations, PharmaEssentia Corporation, Taipei, Taiwan
| |
Collapse
|
17
|
Cheung AHK, Hui CHL, Wong KY, Liu X, Chen B, Kang W, To KF. Out of the cycle: Impact of cell cycle aberrations on cancer metabolism and metastasis. Int J Cancer 2023; 152:1510-1525. [PMID: 36093588 DOI: 10.1002/ijc.34288] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
The use of cell cycle inhibitors has necessitated a better understanding of the cell cycle in tumor biology to optimize the therapeutic approach. Cell cycle aberrations are common in cancers, and it is increasingly acknowledged that these aberrations exert oncogenic effects beyond the cell cycle. Multiple facets such as cancer metabolism, immunity and metastasis are also affected, all of which are beyond the effect of cell proliferation alone. This review comprehensively summarized the important recent findings and advances in these interrelated processes. In cancer metabolism, cell cycle regulators can modulate various pathways in aerobic glycolysis, glucose uptake and gluconeogenesis, mainly through transcriptional regulation and kinase activities. Amino acid metabolism is also regulated through cell cycle progression. On cancer metastasis, metabolic plasticity, immune evasion, tumor microenvironment adaptation and metastatic site colonization are intricately related to the cell cycle, with distinct regulatory mechanisms at each step of invasion and dissemination. Throughout the synthesis of current understanding, knowledge gaps and limitations in the literature are also highlighted, as are new therapeutic approaches such as combinational therapy and challenges in tackling emerging targeted therapy resistance. A greater understanding of how the cell cycle modulates diverse aspects of cancer biology can hopefully shed light on identifying new molecular targets by harnessing the vast potential of the cell cycle.
Collapse
Affiliation(s)
- Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Chris Ho-Lam Hui
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Kit Yee Wong
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoli Liu
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Bonan Chen
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China.,State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Brauer E, Lange T, Keller D, Görlitz S, Cho S, Keye J, Gossen M, Petersen A, Kornak U. Dissecting the influence of cellular senescence on cell mechanics and extracellular matrix formation in vitro. Aging Cell 2023; 22:e13744. [PMID: 36514868 PMCID: PMC10014055 DOI: 10.1111/acel.13744] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022] Open
Abstract
Tissue formation and healing both require cell proliferation and migration, but also extracellular matrix production and tensioning. In addition to restricting proliferation of damaged cells, increasing evidence suggests that cellular senescence also has distinct modulatory effects during wound healing and fibrosis. Yet, a direct role of senescent cells during tissue formation beyond paracrine signaling remains unknown. We here report how individual modules of the senescence program differentially influence cell mechanics and ECM expression with relevance for tissue formation. We compared DNA damage-mediated and DNA damage-independent senescence which was achieved through over-expression of either p16Ink4a or p21Cip1 cyclin-dependent kinase inhibitors in primary human skin fibroblasts. Cellular senescence modulated focal adhesion size and composition. All senescent cells exhibited increased single cell forces which led to an increase in tissue stiffness and contraction in an in vitro 3D tissue formation model selectively for p16 and p21-overexpressing cells. The mechanical component was complemented by an altered expression profile of ECM-related genes including collagens, lysyl oxidases, and MMPs. We found that particularly the lack of collagen and lysyl oxidase expression in the case of DNA damage-mediated senescence foiled their intrinsic mechanical potential. These observations highlight the active mechanical role of cellular senescence during tissue formation as well as the need to synthesize a functional ECM network capable of transferring and storing cellular forces.
Collapse
Affiliation(s)
- Erik Brauer
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Lange
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Keller
- Institute for Medical Genetics and Human Genetics, Charité - Universtitätsmedizin Berlin, Berlin, Germany
| | - Sophie Görlitz
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simone Cho
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jacqueline Keye
- Flow & Mass Cytometry Core Facility, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Manfred Gossen
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Ansgar Petersen
- Julius Wolff Institute, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Kornak
- Institute for Medical Genetics and Human Genetics, Charité - Universtitätsmedizin Berlin, Berlin, Germany.,BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
19
|
Maheshwari M, Yadav N, Hasanain M, Pandey P, Sahai R, Choyal K, Singh A, Nengroo MA, Saini KK, Kumar D, Mitra K, Datta D, Sarkar J. Inhibition of p21 activates Akt kinase to trigger ROS-induced autophagy and impacts on tumor growth rate. Cell Death Dis 2022; 13:1045. [PMID: 36522339 PMCID: PMC9755229 DOI: 10.1038/s41419-022-05486-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Owing to its ability to induce cellular senescence, inhibit PCNA, and arrest cell division cycle by negatively regulating CDKs as well as being a primary target of p53, p21 is traditionally considered a tumor suppressor. Nonetheless, several reports in recent years demonstrated its pro-oncogenic activities such as apoptosis inhibition by cytosolic p21, stimulation of cell motility, and promoting assembly of cyclin D-CDK4/6 complex. These opposing effects of p21 on cell proliferation, supported by the observations of its inconsistent expression in human cancers, led to the emergence of the concept of "antagonistic duality" of p21 in cancer progression. Here we demonstrate that p21 negatively regulates basal autophagy at physiological concentration. Akt activation, upon p21 attenuation, driven ROS accumulation appears to be the major underlying mechanism in p21-mediated modulation of autophagy. We also find p21, as a physiological inhibitor of autophagy, to have oncogenic activity during early events of tumor development while its inhibition favors survival and growth of cancer cells in the established tumor. Our data, thereby, reveal the potential role of autophagy in antagonistic functional duality of p21 in cancer.
Collapse
Affiliation(s)
- Mayank Maheshwari
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Nisha Yadav
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Mohammad Hasanain
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Praveen Pandey
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Rohit Sahai
- grid.418363.b0000 0004 0506 6543Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Kuldeep Choyal
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Akhilesh Singh
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Mushtaq A. Nengroo
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Krishan K. Saini
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Deepak Kumar
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Kalyan Mitra
- grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India ,grid.418363.b0000 0004 0506 6543Electron Microscopy Unit, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India
| | - Dipak Datta
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| | - Jayanta Sarkar
- grid.418363.b0000 0004 0506 6543Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh India ,grid.469887.c0000 0004 7744 2771Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002 India
| |
Collapse
|
20
|
Cho JM, Ghosh R, Mookherjee S, Boudina S, Symons JD. Reduce, Reuse, Recycle, Run ! : 4 Rs to improve cardiac health in advanced age. Aging (Albany NY) 2022; 14:9388-9392. [PMID: 36470665 PMCID: PMC9792203 DOI: 10.18632/aging.204415] [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: 06/02/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
During the aging process damaged/dysfunctional proteins and organelles accumulate and contribute to organ dysfunction. Luckily, there is a conserved intracellular process to reuse and recycle these dysregulated cellular components termed macroautophagy (autophagy). Unfortunately, strong evidence indicates autophagy is compromised with aging, protein quality control is jeopardized, and resultant proteotoxicity can contribute significantly to age-associated organ dysfunction. Are there interventions that can re-establish autophagic flux that is otherwise impaired with aging? With particular regard to the heart, here we review evidence that caloric-restriction, the polyamine spermidine, and the mTOR inhibitor rapamycin, even when initiated late-in-life, restore cardiomyocyte autophagy to an extent that lessens age-associated cardiac dysfunction. Cho et al. provide a physiological intervention to this list i.e., regular physical exercise initiated late-in-life boosts cardiomyocyte autophagic flux and rejuvenates cardiac function in male mice. While this study provides strong evidence for a mechanism whereby heightened physical activity can lead to improved heart health in the context of aging, (i) only male mice were studied; (ii) the intensity of exercise-training might not be suitable for all; and (iii) mice with aging-associated comorbidities were not investigated. Nonetheless, Cho et al. provide robust evidence that a low-cost and simple behavioral intervention initiated late-in-life improves cardiomyocyte autophagic flux and rejuvenates cardiac function.
Collapse
Affiliation(s)
- Jae Min Cho
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Rajeshwary Ghosh
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sohom Mookherjee
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA,Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - J. David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA,Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
21
|
Monavarian M, Elhaw AT, Tang PW, Javed Z, Shonibare Z, Scalise CB, Arend R, Jolly MK, Sewell-Loftin MK, Hempel N, Mythreye K. Emerging perspectives on growth factor metabolic relationships in the ovarian cancer ascites environment. Semin Cancer Biol 2022; 86:709-719. [PMID: 35259492 PMCID: PMC9441472 DOI: 10.1016/j.semcancer.2022.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/07/2023]
Abstract
The ascites ecosystem in ovarian cancer is inhabited by complex cell types and is bathed in an environment rich in cytokines, chemokines, and growth factors that directly and indirectly impact metabolism of cancer cells and tumor associated cells. This milieu of malignant ascites, provides a 'rich' environment for the disease to thrive, contributing to every aspect of advanced ovarian cancer, a devastating gynecological cancer with a significant gap in targeted therapeutics. In this perspective we focus our discussions on the 'acellular' constituents of this liquid malignant tumor microenvironment, and how they influence metabolic pathways. Growth factors, chemokines and cytokines are known modulators of metabolism and have been shown to impact nutrient uptake and metabolic flexibility of tumors, yet few studies have explored how their enrichment in malignant ascites of ovarian cancer patients contributes to the metabolic requirements of ascites-resident cells. We focus here on TGF-βs, VEGF and ILs, which are frequently elevated in ovarian cancer ascites and have all been described to have direct or indirect effects on metabolism, often through gene regulation of metabolic enzymes. We summarize what is known, describe gaps in knowledge, and provide examples from other tumor types to infer potential unexplored roles and mechanisms for ovarian cancer. The distribution and variation in acellular ascites components between patients poses both a challenge and opportunity to further understand how the ascites may contribute to disease heterogeneity. The review also highlights opportunities for studies on ascites-derived factors in regulating the ascites metabolic environment that could act as a unique signature in aiding clinical decisions in the future.
Collapse
Affiliation(s)
- Mehri Monavarian
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA
| | - Amal Taher Elhaw
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Priscilla W Tang
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Zaineb Javed
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA
| | - Zainab Shonibare
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA
| | - Carly Bess Scalise
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Rebecca Arend
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Mohit Kumar Jolly
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, O'Neal Comprehensive Cancer Center, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Nadine Hempel
- Division of Hematology Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh PA 15213, USA.
| | - Karthikeyan Mythreye
- Division of Molecular Cellular Pathology, Department of Pathology, O'Neal Comprehensive Cancer Center, University of Alabama Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
22
|
Was N, Sauer M, Fischer U, Becker M. lncRNA Malat1 and miR-26 cooperate in the regulation of neuronal progenitor cell proliferation and differentiation. RNA (NEW YORK, N.Y.) 2022; 29:rna.079436.122. [PMID: 36302652 PMCID: PMC9808573 DOI: 10.1261/rna.079436.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/19/2022] [Indexed: 06/16/2023]
Abstract
Neurogenesis is a finely tuned process, which depends on the balanced execution of expression programs that regulate cellular differentiation and proliferation. Different molecular players ranging from transcription factors to chromatin modulators control these programs. Adding to the complexity, also non-coding (nc)RNAs take part in this process. Here we analyzed the function of the long non-coding (lnc)RNA Malat1 during neural embryonic stem cell (ESC) differentiation. We find that deletion of Malat1 leads to inhibition of proliferation of neural progenitor cells (NPCs). Interestingly, this co-insides with an increase in the expression of miR-26 family members miR-26a and miR-26b in differentiating ESCs. Inactivation of miR-26a/b rescues the proliferative phenotype of Malat1 knockout (KO) cells and leads to accelerated neuronal differentiation of compound Malat1KO/mir-26KO ESCs. Together our work identifies a so far unknown interaction between Malat1 and miR-26 in the regulation of NPC proliferation and neuronal differentiation.
Collapse
|
23
|
Dose-time and Source-dependent Analysis of Intraoperative Radiotherapy-treated Seroma on Breast Cancer Cell Types: A Pilot Study. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2022. [DOI: 10.5812/ijcm-99862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Intraoperative radiation therapy (IORT) is a novel approach to breast cancer (BC) treatment. Objectives: In this study, we compared the cellular and molecular effects of IORT-treated post-lumpectomy wound fluid (seroma) at the point of IOeRT versus IOxRT on the BC cell line. Methods: Immortalized human BC cell lines: MCF-7, MDA-MB-231, and MCF10 were incubated with seroma from 3 groups of patients (as a pilot study). The first group received Intraoperative electron radiation therapy (IOeRT, Boost dose=12Gy), the second one received IOeRT (Radical dose=21Gy), and the third group was prescribed Intraoperative x-ray radiation therapy (IOxRT, X-ray=20Gy). Cellular and molecular tests were used to investigate how cells are influenced by the IORT-treated seroma. Results: We evaluated the effects of dose-time and source-dependent IORT-treated seroma on BC cell lines. In this study, we observed that IOxRT-treated seroma has the most significant effects on the reduction of proliferation, induced cell cycle arrest, and apoptosis. Furthermore, inhibited migration and invasion of BC cell lines were compared to IOeRT -treated seroma. Conclusions: Although this is a pilot study, we suggest that at 24 h, the IORT (specifically IOxRT)-treated seroma may play an important protective role in the breast tumor bed, which is followed by local recurrence decreases.
Collapse
|
24
|
Lee Y, Kang JS, Ham OJ, Son MY, Lee MO. Gut metabolite trimethylamine N-oxide induces aging-associated phenotype of midbrain organoids for the induced pluripotent stem cell-based modeling of late-onset disease. Front Aging Neurosci 2022; 14:925227. [PMID: 36051303 PMCID: PMC9426463 DOI: 10.3389/fnagi.2022.925227] [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: 04/21/2022] [Accepted: 07/18/2022] [Indexed: 12/01/2022] Open
Abstract
Brain organoids are valuable research models for human development and disease since they mimic the various cell compositions and structures of the human brain; however, they have challenges in presenting aging phenotypes for degenerative diseases. This study analyzed the association between aging and the gut metabolite trimethylamine N-oxide (TMAO), which is highly found in the midbrain of elderly and Parkinson’s disease (PD) patients. TMAO treatment in midbrain organoid induced aging-associated molecular changes, including increased senescence marker expression (P21, P16), p53 accumulation, and epigenetic alterations. In addition, TMAO-treated midbrain organoids have shown parts of neurodegeneration phenotypes, including impaired brain-derived neurotrophic factor (BDNF) signaling, loss of dopaminergic neurons, astrocyte activation, and neuromelanin accumulation. Moreover, we found TMAO treatment-induced pathophysiological phosphorylation of α-synuclein protein at Ser-129 residues and Tau protein at Ser202/Thr205. These results suggest a role of TMAO in the aging and pathogenesis of the midbrain and provide insight into how intestinal dysfunction increases the risk of PD. Furthermore, this system can be utilized as a novel aging model for induced pluripotent stem cell (iPSC)-based modeling of late-onset diseases.
Collapse
Affiliation(s)
- Youngsun Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - Ji Su Kang
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
| | - On-Ju Ham
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Mi-Young Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
- Mi-Young Son,
| | - Mi-Ok Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
- Department of Bioscience, Korea University of Science and Technology (UST), Daejeon, South Korea
- *Correspondence: Mi-Ok Lee,
| |
Collapse
|
25
|
Gillson J, Abd El-Aziz YS, Leck LYW, Jansson PJ, Pavlakis N, Samra JS, Mittal A, Sahni S. Autophagy: A Key Player in Pancreatic Cancer Progression and a Potential Drug Target. Cancers (Basel) 2022; 14:3528. [PMID: 35884592 PMCID: PMC9315706 DOI: 10.3390/cancers14143528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Pancreatic cancer is known to have the lowest survival outcomes among all major cancers, and unfortunately, this has only been marginally improved over last four decades. The innate characteristics of pancreatic cancer include an aggressive and fast-growing nature from powerful driver mutations, a highly defensive tumor microenvironment and the upregulation of advantageous survival pathways such as autophagy. Autophagy involves targeted degradation of proteins and organelles to provide a secondary source of cellular supplies to maintain cell growth. Elevated autophagic activity in pancreatic cancer is recognized as a major survival pathway as it provides a plethora of support for tumors by supplying vital resources, maintaining tumour survival under the stressful microenvironment and promoting other pathways involved in tumour progression and metastasis. The combination of these features is unique to pancreatic cancer and present significant resistance to chemotherapeutic strategies, thus, indicating a need for further investigation into therapies targeting this crucial pathway. This review will outline the autophagy pathway and its regulation, in addition to the genetic landscape and tumor microenvironment that contribute to pancreatic cancer severity. Moreover, this review will also discuss the mechanisms of novel therapeutic strategies that inhibit autophagy and how they could be used to suppress tumor progression.
Collapse
Affiliation(s)
- Josef Gillson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Yomna S. Abd El-Aziz
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta 31527, Egypt
| | - Lionel Y. W. Leck
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Patric J. Jansson
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Cancer Drug Resistance and Stem Cell Program, University of Sydney, Sydney, NSW 2006, Australia
| | - Nick Pavlakis
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
| | - Jaswinder S. Samra
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| | - Anubhav Mittal
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Upper GI Surgical Unit, Royal North Shore Hospital and North Shore Private Hospital, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
- School of Medicine, University of Notre Dame, Darlinghurst, Sydney, NSW 2010, Australia
| | - Sumit Sahni
- Faculty of Medicine and Health, University of Sydney, Camperdown, Sydney, NSW 2050, Australia; (J.G.); (Y.S.A.E.-A.); (L.Y.W.L.); (P.J.J.); (N.P.); (J.S.S.); (A.M.)
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, St Leonards, Sydney, NSW 2065, Australia
- Australian Pancreatic Centre, St Leonards, Sydney, NSW 2065, Australia
| |
Collapse
|
26
|
Guo Y, Chomiak A, Hong Y, Lowe CC, Kopsidas CA, Chan WC, Andrade J, Pan H, Zhou X, Monuki ES, Feng Y. Histone H2A ubiquitination resulting from Brap loss of function connects multiple aging hallmarks and accelerates neurodegeneration. iScience 2022; 25:104519. [PMID: 35754718 PMCID: PMC9213774 DOI: 10.1016/j.isci.2022.104519] [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: 10/05/2021] [Revised: 04/06/2022] [Accepted: 05/29/2022] [Indexed: 01/10/2023] Open
Abstract
Aging is an intricate process characterized by multiple hallmarks including stem cell exhaustion, genome instability, epigenome alteration, impaired proteostasis, and cellular senescence. Whereas each of these traits is detrimental at the cellular level, it remains unclear how they are interconnected to cause systemic organ deterioration. Here we show that abrogating Brap, a BRCA1-associated protein essential for neurogenesis, results in persistent DNA double-strand breaks and elevation of histone H2A mono- and poly-ubiquitination (H2Aub). These defects extend to cellular senescence and proteasome-mediated histone H2A proteolysis with alterations in cells' proteomic and epigenetic states. Brap deletion in the mouse brain causes neuroinflammation, impaired proteostasis, accelerated neurodegeneration, and substantially shortened the lifespan. We further show the elevation of H2Aub also occurs in human brain tissues with Alzheimer's disease. These data together suggest that chromatin aberrations mediated by H2Aub may act as a nexus of multiple aging hallmarks and promote tissue-wide degeneration.
Collapse
Affiliation(s)
- Yan Guo
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Alison.A. Chomiak
- Department of Neurology, Northwestern University Feinberg School of Medicine, 303 E. Superior Street, Chicago, IL 60611, USA
| | - Ye Hong
- University of Turku, Turku 20500, Finland
| | - Clara C. Lowe
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Caroline A. Kopsidas
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Wen-Ching Chan
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Jorge Andrade
- Center for Research Informatics, University of Chicago, Chicago, IL 60637, USA
| | - Hongna Pan
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Xiaoming Zhou
- Department of Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| | - Edwin S. Monuki
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Yuanyi Feng
- Department of Biochemistry and Molecular Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
| |
Collapse
|
27
|
Abstract
Senescence is a cellular response to a variety of stress signals that is characterized by a stable withdrawal from the cell cycle and major changes in cell morphology and physiology. While most research on senescence has been performed on non-cancer cells, it is evident that cancer cells can also mount a senescence response. In this Review, we discuss how senescence can be induced in cancer cells. We describe the distinctive features of senescent cancer cells and how these changes in cellular physiology might be exploited for the selective eradication of these cells (senolysis). We discuss activation of the host immune system as a particularly attractive way to clear senescent cancer cells. Finally, we consider the challenges and opportunities provided by a 'one-two punch' sequential treatment of cancer with pro-senescence therapy followed by senolytic therapy.
Collapse
Affiliation(s)
- Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lina Lankhorst
- Cancer, Stem Cells & Developmental Biology programme, Utrecht University, Utrecht, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
28
|
Singh G, Liu P, Yao KR, Strasser JM, Hlynialuk C, Leinonen-Wright K, Teravskis PJ, Choquette JM, Ikramuddin J, Bresinsky M, Nelson KM, Liao D, Ashe KH, Walters MA, Pockes S. Caspase-2 Inhibitor Blocks Tau Truncation and Restores Excitatory Neurotransmission in Neurons Modeling FTDP-17 Tauopathy. ACS Chem Neurosci 2022; 13:1549-1557. [PMID: 35522720 DOI: 10.1021/acschemneuro.2c00100] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Synaptic and cognitive deficits mediated by a severe reduction in excitatory neurotransmission caused by a disproportionate accumulation of the neuronal protein tau in dendritic spines is a fundamental mechanism that has been found repeatedly in models of tauopathies, including Alzheimer's disease, Lewy body dementia, frontotemporal dementia, and traumatic brain injury. Synapses thus damaged may contribute to dementia, among the most feared cause of debilitation in the elderly, and currently there are no treatments to repair them. Caspase-2 (Casp2) is an essential component of this pathological cascade. Although it is believed that Casp2 exerts its effects by hydrolyzing tau at aspartate-314, forming Δtau314, it is also possible that a noncatalytic mechanism is involved because catalytically dead Casp2 is biologically active in at least one relevant cellular pathway, that is, autophagy. To decipher whether the pathological effects of Casp2 on synaptic function are due to its catalytic or noncatalytic properties, we discovered and characterized a new Casp2 inhibitor, compound 1 [pKi (Casp2) = 8.12], which is 123-fold selective versus Casp3 and >2000-fold selective versus Casp1, Casp6, Casp7, and Casp9. In an in vitro assay based on Casp2-mediated cleavage of tau, compound 1 blocked the production of Δtau314. Importantly, compound 1 prevented tau from accumulating excessively in dendritic spines and rescued excitatory neurotransmission in cultured primary rat hippocampal neurons expressing the P301S tau variant linked to FTDP-17, a familial tauopathy. These results support the further development of small-molecule Casp2 inhibitors to treat synaptic deficits in tauopathies.
Collapse
Affiliation(s)
- Gurpreet Singh
- Department of Medicinal Chemistry, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Peng Liu
- Department of Neurology, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Katherine R. Yao
- Department of Neuroscience, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jessica M. Strasser
- Department of Medicinal Chemistry, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Chris Hlynialuk
- Department of Neurology, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Kailee Leinonen-Wright
- Department of Neurology, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Peter J. Teravskis
- Department of Neuroscience, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jessica M. Choquette
- Department of Neuroscience, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Junaid Ikramuddin
- Department of Neuroscience, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Merlin Bresinsky
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Kathryn M. Nelson
- Department of Medicinal Chemistry, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Dezhi Liao
- Department of Neuroscience, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Karen H. Ashe
- Department of Neurology, The University of Minnesota, Minneapolis, Minnesota 55455, United States
- Veterans Administration Medical Center, GRECC, Minneapolis, Minnesota 55417, United States
| | - Michael A. Walters
- Department of Medicinal Chemistry, The University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Steffen Pockes
- Department of Medicinal Chemistry, The University of Minnesota, Minneapolis, Minnesota 55455, United States
- Department of Neurology, The University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute of Pharmacy, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| |
Collapse
|
29
|
Long-term consumption of green tea EGCG enhances murine healthspan by mitigating multiple aspects of cellular senescence in mitotic and post-mitotic tissues, gut dysbiosis, and immunosenescence. J Nutr Biochem 2022; 107:109068. [DOI: 10.1016/j.jnutbio.2022.109068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
|
30
|
Liu J, Zhang J, Lin X, Boyce BF, Zhang H, Xing L. Age-associated callus senescent cells produce TGF-β1 that inhibits fracture healing in aged mice. J Clin Invest 2022; 132:e148073. [PMID: 35426372 PMCID: PMC9012290 DOI: 10.1172/jci148073] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 02/16/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence plays an important role in human diseases, including osteoporosis and osteoarthritis. Senescent cells (SCs) produce the senescence-associated secretory phenotype to affect the function of neighboring cells and SCs themselves. Delayed fracture healing is common in the elderly and is accompanied by reduced mesenchymal progenitor cells (MPCs). However, the contribution of cellular senescence to fracture healing in the aged has not to our knowledge been studied. Here, we used C57BL/6J 4-month-old young and 20-month-old aged mice and demonstrated a rapid increase in SCs in the fracture callus of aged mice. The senolytic drugs dasatinib plus quercetin enhanced fracture healing in aged mice. Aged callus SCs inhibited the growth and proliferation of callus-derived MPCs (CaMPCs) and expressed high levels of TGF-β1. TGF-β-neutralizing Ab prevented the inhibitory effects of aged callus SCs on CaMPCs and promoted fracture healing in aged mice, which was associated with increased CaMPCs and proliferating cells. Thus, fracture triggered a significant cellular senescence in the callus cells of aged mice, which inhibited MPCs by expressing TGF-β1. Short-term administration of dasatinib plus quercetin depleted callus SCs and accelerated fracture healing in aged mice. Senolytic drugs represent a promising therapy, while TGF-β1 signaling is a molecular mechanism for fractures in the elderly via SCs.
Collapse
Affiliation(s)
- Jiatong Liu
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Jun Zhang
- Plastic Surgery Center, Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Xi Lin
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Brendan F. Boyce
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Hengwei Zhang
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
31
|
p27, The Cell Cycle and Alzheimer´s Disease. Int J Mol Sci 2022; 23:ijms23031211. [PMID: 35163135 PMCID: PMC8835212 DOI: 10.3390/ijms23031211] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
The cell cycle consists of successive events that lead to the generation of new cells. The cell cycle is regulated by different cyclins, cyclin-dependent kinases (CDKs) and their inhibitors, such as p27Kip1. At the nuclear level, p27Kip1 has the ability to control the evolution of different phases of the cell cycle and oppose cell cycle progression by binding to CDKs. In the cytoplasm, diverse functions have been described for p27Kip1, including microtubule remodeling, axonal transport and phagocytosis. In Alzheimer’s disease (AD), alterations to cycle events and a purported increase in neurogenesis have been described in the early disease process before significant pathological changes could be detected. However, most neurons cannot progress to complete their cell division and undergo apoptotic cell death. Increased levels of both the p27Kip1 levels and phosphorylation status have been described in AD. Increased levels of Aβ42, tau hyperphosphorylation or even altered insulin signals could lead to alterations in p27Kip1 post-transcriptional modifications, causing a disbalance between the levels and functions of p27Kip1 in the cytoplasm and nucleus, thus inducing an aberrant cell cycle re-entry and alteration of extra cell cycle functions. Further studies are needed to completely understand the role of p27Kip1 in AD and the therapeutic opportunities associated with the modulation of this target.
Collapse
|
32
|
Arora M, Bogenberger JM, Abdelrahman AM, Yonkus J, Alva-Ruiz R, Leiting JL, Chen X, Serrano Uson Junior PL, Dumbauld CR, Baker AT, Gamb SI, Egan JB, Zhou Y, Nagalo BM, Meurice N, Eskelinen EL, Salomao MA, Kosiorek HE, Braggio E, Barrett MT, Buetow KH, Sonbol MB, Mansfield AS, Roberts LR, Bekaii-Saab TS, Ahn DH, Truty MJ, Borad MJ. Synergistic combination of cytotoxic chemotherapy and cyclin-dependent kinase 4/6 inhibitors in biliary tract cancers. Hepatology 2022; 75:43-58. [PMID: 34407567 DOI: 10.1002/hep.32102] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Biliary tract cancers (BTCs) are uncommon, but highly lethal, gastrointestinal malignancies. Gemcitabine/cisplatin is a standard-of-care systemic therapy, but has a modest impact on survival and harbors toxicities, including myelosuppression, nephropathy, neuropathy, and ototoxicity. Whereas BTCs are characterized by aberrations activating the cyclinD1/cyclin-dependent kinase (CDK)4/6/CDK inhibitor 2a/retinoblastoma pathway, clinical use of CDK4/6 inhibitors as monotherapy is limited by lack of validated biomarkers, diffident preclinical efficacy, and development of acquired drug resistance. Emerging studies have explored therapeutic strategies to enhance the antitumor efficacy of CDK4/6 inhibitors by the combination with chemotherapy regimens, but their mechanism of action remains elusive. APPROACH AND RESULTS Here, we report in vitro and in vivo synergy in BTC models, showing enhanced efficacy, reduced toxicity, and better survival with a combination comprising gemcitabine/cisplatin and CDK4/6 inhibitors. Furthermore, we demonstrated that abemaciclib monotherapy had only modest efficacy attributable to autophagy-induced resistance. Notably, triplet therapy was able to potentiate efficacy through elimination of the autophagic flux. Correspondingly, abemaciclib potentiated ribonucleotide reductase catalytic subunit M1 reduction, resulting in sensitization to gemcitabine. CONCLUSIONS As such, these data provide robust preclinical mechanistic evidence of synergy between gemcitabine/cisplatin and CDK4/6 inhibitors and delineate a path forward for translation of these findings to preliminary clinical studies in advanced BTC patients.
Collapse
Affiliation(s)
- Mansi Arora
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - James M Bogenberger
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Jennifer Yonkus
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Xianfeng Chen
- Department of Informatics, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Chelsae R Dumbauld
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Alexander T Baker
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott I Gamb
- Microscopy and Cell Analysis Core, Mayo Clinic, Rochester, Minnesota, USA
| | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yumei Zhou
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Bolni Marius Nagalo
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nathalie Meurice
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Marcela A Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Heidi E Kosiorek
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona, USA
| | - Esteban Braggio
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Michael T Barrett
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Kenneth H Buetow
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Mohamad B Sonbol
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Aaron S Mansfield
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lewis R Roberts
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tanios S Bekaii-Saab
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Daniel H Ahn
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitesh J Borad
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
33
|
Ziegler DV, Huber K, Fajas L. The Intricate Interplay between Cell Cycle Regulators and Autophagy in Cancer. Cancers (Basel) 2021; 14:cancers14010153. [PMID: 35008317 PMCID: PMC8750274 DOI: 10.3390/cancers14010153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary Autophagy is an intracellular catabolic program regulated by multiple external and internal cues. A large amount of evidence unraveled that cell-cycle regulators are crucial in its control. This review highlights the interplay between cell-cycle regulators, including cyclin-dependent kinase inhibitors, cyclin-dependent kinases, and E2F factors, in the control of autophagy all along the cell cycle. Beyond the intimate link between cell cycle and autophagy, this review opens therapeutic perspectives in modulating together these two aspects to block cancer progression. Abstract In the past decade, cell cycle regulators have extended their canonical role in cell cycle progression to the regulation of various cellular processes, including cellular metabolism. The regulation of metabolism is intimately connected with the function of autophagy, a catabolic process that promotes the efficient recycling of endogenous components from both extrinsic stress, e.g., nutrient deprivation, and intrinsic sub-lethal damage. Mediating cellular homeostasis and cytoprotection, autophagy is found to be dysregulated in numerous pathophysiological contexts, such as cancer. As an adaptative advantage, the upregulation of autophagy allows tumor cells to integrate stress signals, escaping multiple cell death mechanisms. Nevertheless, the precise role of autophagy during tumor development and progression remains highly context-dependent. Recently, multiple articles has suggested the importance of various cell cycle regulators in the modulation of autophagic processes. Here, we review the current clues indicating that cell-cycle regulators, including cyclin-dependent kinase inhibitors (CKIs), cyclin-dependent kinases (CDKs), and E2F transcription factors, are intrinsically linked to the regulation of autophagy. As an increasing number of studies highlight the importance of autophagy in cancer progression, we finally evoke new perspectives in therapeutic avenues that may include both cell cycle inhibitors and autophagy modulators to synergize antitumor efficacy.
Collapse
|
34
|
Eng GWL, Zheng Y, Yap DWT, Teo AYT, Cheong JK. Autophagy and ncRNAs: Dangerous Liaisons in the Crosstalk between the Tumor and Its Microenvironment. Cancers (Basel) 2021; 14:cancers14010020. [PMID: 35008183 PMCID: PMC8750064 DOI: 10.3390/cancers14010020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Tumor cells communicate with the stromal cells within the tumor microenvironment (TME) to create a conducive environment for tumor growth. One major avenue for mediating crosstalk between various cell types in the TME involves exchanges of molecular payloads in the form of extracellular vesicles/exosomes. Autophagy is a fundamental mechanism to maintain intracellular homeostasis but recent reports suggest that secretory autophagy plays an important role in promoting secretion of exosomes that are packaged with non-coding RNAs (ncRNAs) and other biomolecules from the donor cell. Uptake of exosomal autophagy-modulating ncRNAs by recipient cells may further perpetuate tumor progression. Abstract Autophagy is a fundamental cellular homeostasis mechanism known to play multifaceted roles in the natural history of cancers over time. It has recently been shown that autophagy also mediates the crosstalk between the tumor and its microenvironment by promoting the export of molecular payloads such as non-coding RNA (ncRNAs) via LC3-dependent Extracellular Vesicle loading and secretion (LDELS). In turn, the dynamic exchange of exosomal ncRNAs regulate autophagic responses in the recipient cells within the tumor microenvironment (TME), for both tumor and stromal cells. Autophagy-dependent phenotypic changes in the recipient cells further enhance tumor growth and metastasis, through diverse biological processes, including nutrient supplementation, immune evasion, angiogenesis, and therapeutic resistance. In this review, we discuss how the feedforward autophagy-ncRNA axis orchestrates vital communications between various cell types within the TME ecosystem to promote cancer progression.
Collapse
Affiliation(s)
- Gracie Wee Ling Eng
- Precision Medicine Programme, Yong Loo Lin School of Medicine (YLLSoM), National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228, Singapore; (G.W.L.E.); (Y.Z.); (D.W.T.Y.); (A.Y.T.T.)
| | - Yilong Zheng
- Precision Medicine Programme, Yong Loo Lin School of Medicine (YLLSoM), National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228, Singapore; (G.W.L.E.); (Y.Z.); (D.W.T.Y.); (A.Y.T.T.)
| | - Dominic Wei Ting Yap
- Precision Medicine Programme, Yong Loo Lin School of Medicine (YLLSoM), National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228, Singapore; (G.W.L.E.); (Y.Z.); (D.W.T.Y.); (A.Y.T.T.)
| | - Andrea York Tiang Teo
- Precision Medicine Programme, Yong Loo Lin School of Medicine (YLLSoM), National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228, Singapore; (G.W.L.E.); (Y.Z.); (D.W.T.Y.); (A.Y.T.T.)
| | - Jit Kong Cheong
- Precision Medicine Programme, Yong Loo Lin School of Medicine (YLLSoM), National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228, Singapore; (G.W.L.E.); (Y.Z.); (D.W.T.Y.); (A.Y.T.T.)
- NUS Centre for Cancer Research, National University of Singapore, 14 Medical Dr, Centre for Translational Medicine #12-01, Singapore 117599, Singapore
- Department of Biochemistry, YLLSoM, National University of Singapore, 8 Medical Drive, MD7 #03-09, Singapore 117597, Singapore
- Correspondence: ; Tel.: +65-66016388
| |
Collapse
|
35
|
Kudlova N, Slavik H, Duskova P, Furst T, Srovnal J, Bartek J, Mistrik M, Hajduch M. An efficient, non-invasive approach for in-vivo sampling of hair follicles: design and applications in monitoring DNA damage and aging. Aging (Albany NY) 2021; 13:25004-25024. [PMID: 34874896 PMCID: PMC8714131 DOI: 10.18632/aging.203744] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023]
Abstract
In accordance with the 3 Rs principle (to replace, reduce and refine) animal models in biomedical research, we have developed and applied a new approach for sampling and analyzing hair follicles in various experimental settings. This involves use of a convenient device for non-invasive collection of hair follicles and processing methods that provide sufficient amounts of biological material to replace stressful and painful biopsies. Moreover, the main components of hair follicles are live cells of epithelial origin, which are highly relevant for most types of malignant tumors, so they provide opportunities for studying aging-related pathologies including cancer. Here, we report the successful use of the method to obtain mouse hair follicular cells for genotyping, quantitative PCR, and quantitative immunofluorescence. We present proof of concept data demonstrating its utility for routine genotyping and monitoring changes in quality and expression levels of selected proteins in mice after gamma irradiation and during natural or experimentally induced aging. We also performed pilot translation of animal experiments to human hair follicles irradiated ex vivo. Our results highlight the value of hair follicles as biological material for convenient in vivo sampling and processing in both translational research and routine applications, with a broad range of ethical and logistic advantages over currently used biopsy-based approaches.
Collapse
Affiliation(s)
- Natalie Kudlova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Hanus Slavik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Pavlina Duskova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Tomas Furst
- Faculty of Science, Palacky University and University Hospital in Olomouc, Olomouc 779 00, Czech Republic
| | - Josef Srovnal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Jiri Bartek
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic.,Danish Cancer Society Research Center, Copenhagen DK-2100, Denmark.,Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm 171 77, Sweden
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University and University Hospital Olomouc, Olomouc 779 00, Czech Republic
| |
Collapse
|
36
|
Bhurta D, Bharate SB. Analyzing the scaffold diversity of cyclin-dependent kinase inhibitors and revisiting the clinical and preclinical pipeline. Med Res Rev 2021; 42:654-709. [PMID: 34605036 DOI: 10.1002/med.21856] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 07/04/2021] [Accepted: 09/21/2021] [Indexed: 12/17/2022]
Abstract
Kinases have gained an important place in the list of vital therapeutic targets because of their overwhelming clinical success in the last two decades. Among various clinically validated kinases, the cyclin-dependent kinases (CDK) are one of the extensively studied drug targets for clinical development. Food and Drug Administration has approved three CDK inhibitors for therapeutic use, and at least 27 inhibitors are under active clinical development. In the last decade, research and development in this area took a rapid pace, and thus the analysis of scaffold diversity is essential for future drug design. Available reviews lack the systematic study and discussion on the scaffold diversity of CDK inhibitors. Herein we have reviewed and critically analyzed the chemical diversity present in the preclinical and clinical pipeline of CDK inhibitors. Our analysis has shown that although several scaffolds represent CDK inhibitors, only the amino-pyrimidine is a well-represented scaffold. The three-nitrogen framework of amino-pyrimidine is a fundamental hinge-binding unit. Further, we have discussed the selectivity aspects among CDKs, the clinical trial dose-limiting toxicities, and highlighted the most advanced clinical candidates. We also discuss the changing paradigm towards selective inhibitors and an overview of ATP-binding pockets of all druggable CDKs. We carefully analyzed the clinical pipeline to unravel the candidates that are currently under active clinical development. In addition to the plenty of dual CDK4/6 inhibitors, there are many selective CDK7, CDK9, and CDK8/19 inhibitors in the clinical pipeline.
Collapse
Affiliation(s)
- Deendyal Bhurta
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Sandip B Bharate
- Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
37
|
Mehraj U, Ganai RA, Macha MA, Hamid A, Zargar MA, Bhat AA, Nasser MW, Haris M, Batra SK, Alshehri B, Al-Baradie RS, Mir MA, Wani NA. The tumor microenvironment as driver of stemness and therapeutic resistance in breast cancer: New challenges and therapeutic opportunities. Cell Oncol (Dordr) 2021; 44:1209-1229. [PMID: 34528143 DOI: 10.1007/s13402-021-00634-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer (BC), the second most common cause of cancer-related deaths, remains a significant threat to the health and wellness of women worldwide. The tumor microenvironment (TME), comprising cellular components, such as cancer-associated fibroblasts (CAFs), immune cells, endothelial cells and adipocytes, and noncellular components such as extracellular matrix (ECM), has been recognized as a critical contributor to the development and progression of BC. The interplay between TME components and cancer cells promotes phenotypic heterogeneity, cell plasticity and cancer cell stemness that impart tumor dormancy, enhanced invasion and metastasis, and the development of therapeutic resistance. While most previous studies have focused on targeting cancer cells with a dismal prognosis, novel therapies targeting stromal components are currently being evaluated in preclinical and clinical studies, and are already showing improved efficacies. As such, they may offer better means to eliminate the disease effectively. CONCLUSIONS In this review, we focus on the evolving concept of the TME as a key player regulating tumor growth, metastasis, stemness, and the development of therapeutic resistance. Despite significant advances over the last decade, several clinical trials focusing on the TME have failed to demonstrate promising effectiveness in cancer patients. To expedite clinical efficacy of TME-directed therapies, a deeper understanding of the TME is of utmost importance. Secondly, the efficacy of TME-directed therapies when used alone or in combination with chemo- or radiotherapy, and the tumor stage needs to be studied. Likewise, identifying molecular signatures and biomarkers indicating the type of TME will help in determining precise TME-directed therapies.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India
| | - Rais A Ganai
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mohammed A Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Ajaz A Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory of Animal Research, Qatar University, Doha, Qatar
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska, Lincoln, NE, USA.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Raid Saleem Al-Baradie
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Manzoor A Mir
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India.
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India.
| |
Collapse
|
38
|
Chandra Jena B, Sarkar S, Rout L, Mandal M. The transformation of cancer-associated fibroblasts: Current perspectives on the role of TGF-β in CAF mediated tumor progression and therapeutic resistance. Cancer Lett 2021; 520:222-232. [PMID: 34363903 DOI: 10.1016/j.canlet.2021.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022]
Abstract
Over the last few years, the Transforming growth factor- β (TGF-β) has been significantly considered as an effective and ubiquitous mediator of cell growth. The cytokine, TGF-β is being increasingly recognized as the most potent inducer of cancer cell initiation, differentiation, migration as well as progression through both the SMAD-dependent and independent pathways. There is growing evidence that supports the role of secretory cytokine TGF-β as a crucial mediator of tumor-stroma crosstalk. Contextually, the CAFs are the prominent component of tumor stroma that helps in tumor progression and onset of chemoresistance. The interplay between the CAFs and the tumor cells through the paracrine signals is facilitated by cytokine TGF-β to induce the malignant progression. Here in this review, we have dissected the most recent advancements in understanding the mechanisms of TGF-β induced CAF activation, their multiple origins, and most importantly their role in conferring chemoresistance. Considering the pivotal role of TGF-β in tumor perogression and associated stemness, it is one the proven clinical targets We have also included the clinical trials going on, targeting the TGF-β and CAFs crosstalk with the tumor cells. Ultimately, we have underscored some of the outstanding issues that must be deciphered with utmost importance to unravel the successful strategies of anti-cancer therapies.
Collapse
Affiliation(s)
- Bikash Chandra Jena
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Siddik Sarkar
- CSIR-Indian Institue of Chemical Biology, Translational Research Unit of Excellence, Kolkata, West Bengal, India
| | - Lipsa Rout
- Department of Chemistry, Institute of Technical Education and Research, Siksha'O'Anusandhan Deemed to be University, Bhubaneswar, Odisha, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India.
| |
Collapse
|
39
|
Nardone V, Barbarino M, Angrisani A, Correale P, Pastina P, Cappabianca S, Reginelli A, Mutti L, Miracco C, Giannicola R, Giordano A, Pirtoli L. CDK4, CDK6/cyclin-D1 Complex Inhibition and Radiotherapy for Cancer Control: A Role for Autophagy. Int J Mol Sci 2021; 22:8391. [PMID: 34445095 PMCID: PMC8395054 DOI: 10.3390/ijms22168391] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
The expanding clinical application of CDK4- and CDK6-inhibiting drugs in the managements of breast cancer has raised a great interest in testing these drugs in other neoplasms. The potential of combining these drugs with other therapeutic approaches seems to be an interesting work-ground to explore. Even though a potential integration of CDK4 and CDK6 inhibitors with radiotherapy (RT) has been hypothesized, this kind of approach has not been sufficiently pursued, neither in preclinical nor in clinical studies. Similarly, the most recent discoveries focusing on autophagy, as a possible target pathway able to enhance the antitumor efficacy of CDK4 and CDK6 inhibitors is promising but needs more investigations. The aim of this review is to discuss the recent literature on the field in order to infer a rational combination strategy including cyclin-D1/CDK4-CDK6 inhibitors, RT, and/or other anticancer agents targeting G1-S phase cell cycle transition.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Marcella Barbarino
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.)
| | - Antonio Angrisani
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Pierpaolo Correale
- Medical Oncology Unit, Grand Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy; (P.C.); (R.G.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Pierpaolo Pastina
- Section of Radiation Oncology, Medical School, University of Siena, 53100 Siena, Italy;
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (A.A.); (S.C.); (A.R.)
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Clelia Miracco
- Pathological Anatomy Unit, Department of Medical, Surgical and Neurological Science, University of Siena, 53100 Siena, Italy;
| | - Rocco Giannicola
- Medical Oncology Unit, Grand Metropolitan Hospital “Bianchi-Melacrino-Morelli”, 89124 Reggio Calabria, Italy; (P.C.); (R.G.)
| | - Antonio Giordano
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (M.B.); (A.G.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19104, USA; (L.M.); (L.P.)
| |
Collapse
|
40
|
Cayo A, Segovia R, Venturini W, Moore-Carrasco R, Valenzuela C, Brown N. mTOR Activity and Autophagy in Senescent Cells, a Complex Partnership. Int J Mol Sci 2021; 22:ijms22158149. [PMID: 34360912 PMCID: PMC8347619 DOI: 10.3390/ijms22158149] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is a form of proliferative arrest triggered in response to a wide variety of stimuli and characterized by unique changes in cell morphology and function. Although unable to divide, senescent cells remain metabolically active and acquire the ability to produce and secrete bioactive molecules, some of which have recognized pro-inflammatory and/or pro-tumorigenic actions. As expected, this “senescence-associated secretory phenotype (SASP)” accounts for most of the non-cell-autonomous effects of senescent cells, which can be beneficial or detrimental for tissue homeostasis, depending on the context. It is now evident that many features linked to cellular senescence, including the SASP, reflect complex changes in the activities of mTOR and other metabolic pathways. Indeed, the available evidence indicates that mTOR-dependent signaling is required for the maintenance or implementation of different aspects of cellular senescence. Thus, depending on the cell type and biological context, inhibiting mTOR in cells undergoing senescence can reverse senescence, induce quiescence or cell death, or exacerbate some features of senescent cells while inhibiting others. Interestingly, autophagy—a highly regulated catabolic process—is also commonly upregulated in senescent cells. As mTOR activation leads to repression of autophagy in non-senescent cells (mTOR as an upstream regulator of autophagy), the upregulation of autophagy observed in senescent cells must take place in an mTOR-independent manner. Notably, there is evidence that autophagy provides free amino acids that feed the mTOR complex 1 (mTORC1), which in turn is required to initiate the synthesis of SASP components. Therefore, mTOR activation can follow the induction of autophagy in senescent cells (mTOR as a downstream effector of autophagy). These functional connections suggest the existence of autophagy regulatory pathways in senescent cells that differ from those activated in non-senescence contexts. We envision that untangling these functional connections will be key for the generation of combinatorial anti-cancer therapies involving pro-senescence drugs, mTOR inhibitors, and/or autophagy inhibitors.
Collapse
Affiliation(s)
- Angel Cayo
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Raúl Segovia
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Whitney Venturini
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, University of Talca, Talca 346000, Chile;
| | - Rodrigo Moore-Carrasco
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, University of Talca, Talca 346000, Chile;
| | - Claudio Valenzuela
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Nelson Brown
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
- Correspondence:
| |
Collapse
|
41
|
Giri S, Takada A, Paudel D, Yoshida K, Furukawa M, Kuramitsu Y, Matsushita K, Abiko Y, Furuichi Y. An in vitro senescence model of gingival epithelial cell induced by hydrogen peroxide treatment. Odontology 2021; 110:44-53. [PMID: 34143349 DOI: 10.1007/s10266-021-00630-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/14/2021] [Indexed: 11/30/2022]
Abstract
Gingival tissue shows progressive changes with aging and an in vitro model of gingival tissue could be useful in understanding age-associated oral diseases. The present study aims to establish a hydrogen peroxide (H2O2) treatment model to induce aging in human gingival epithelial cells. In addition, fisetin, a flavonoid component studied for the anti-aging property is used to examine if it could reverse the induced senescence. Primary human gingival epithelial progenitor (HGEPp) cells were cultured and treated with different concentrations of H2O2. A cell vitality and morphology, senescence-associated beta-galactosidase (SA-β-gal) staining, mRNA and protein expression analysis of known senescence markers p16, p21, and p53, and cell cycle assay were performed. The cells showed dose-dependent changes in vitality and morphology, SA-β-gal staining, relative mRNA and protein expression, and cell cycle assay after H2O2 treatment. Based on these results, 400 μM H2O2 was considered as an optimal concentration to induce senescence. Treatment of senescence-induced cells with fisetin downregulated all the senescence markers used in this study. In conclusion, a senescence model of gingival epithelial cells induced by hydrogen peroxide treatment was established which could be employed to study age-related periodontal diseases.
Collapse
Affiliation(s)
- Sarita Giri
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, 061-0293, Japan
| | - Ayuko Takada
- Division of Biochemistry, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Durga Paudel
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Koki Yoshida
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Masae Furukawa
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuhiro Kuramitsu
- Research Institute of Cancer Prevention, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Kenji Matsushita
- Department of Oral Disease Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yoshihiro Abiko
- Division of Oral Medicine and Pathology, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Japan
| | - Yasushi Furuichi
- Division of Periodontology and Endodontology, Department of Oral Rehabilitation, School of Dentistry, Health Sciences University of Hokkaido, Ishikari-Tobetsu, 061-0293, Japan.
| |
Collapse
|
42
|
Tokgun O, Tokgun PE, Turel S, Inal B, Inci K, Tan S, Can Alvur O. Bryonia multiflora Extract Induces Autophagy via Regulating Long Non-coding RNAs in Breast Cancer Cells. Nutr Cancer 2021; 73:1792-1803. [PMID: 34024207 DOI: 10.1080/01635581.2021.1922717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Bryonia multiflora, one of the species of Bryonia L. (Cucurbitaceae) genus, is a perennial, dioecious, herbaceous plant with rhizome-shaped roots. Bryonia species have anti-inflammatory, antimicrobial, cytotoxic, antioxidant, etc., activities and their components consume antitumoral effects. Purpose of the study to investigate the effect of Bryonia Multiflora extract (BMST) on breast cancer cells. Our results revealed that MCF-7 and MDA-MB-231 cells underwent significant morphological changes leading to cell rounding. No significant changes were observed in the cell viability by MTT. Acridine orange staining of our cells gave rise to think that BMST might lead our cells to autophagy. Therefore, possible molecular mechanisms underlying morphological changes such as autophagy (LC-3B, Beclin, AMBRA1) and apoptosis (Bcl-2) were evaluated on mRNA and protein levels. BMST treated MCF-7 and MDA-MB-231 cells had increased levels of autophagy markers whereas decreased levels of Bcl-2. p21 levels were also found to be increased in both cells. Analysis of lncRNA expressions has shown that BMST treatment led to changes in the expression levels of several lncRNAs playing roles in autophagy. The current study has shown that BMST induces autophagy in MCF-7 and MDA-MB-231 cells via regulating the lncRNAs revealing that BMST could be a promising therapeutic agent.
Collapse
Affiliation(s)
- Onur Tokgun
- Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey.,Department of Cancer Molecular Biology, Institute of Medical Sciences, Pamukkale University, Denizli, Turkey
| | - Pervin Elvan Tokgun
- Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Samet Turel
- Medical Genetics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Behcet Inal
- Faculty of Agriculture, Department of Agricultural Biotechnology, Siirt University, Siirt, Turkey
| | - Kubilay Inci
- Department of Cancer Molecular Biology, Institute of Medical Sciences, Pamukkale University, Denizli, Turkey
| | - Secil Tan
- Department of Cancer Molecular Biology, Institute of Medical Sciences, Pamukkale University, Denizli, Turkey
| | - Ozge Can Alvur
- Medical Biology, Faculty of Medicine, Yuzuncu Yıl University, Van, Turkey
| |
Collapse
|
43
|
Schwab N, Ju Y, Hazrati LN. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol Commun 2021; 9:82. [PMID: 33964983 PMCID: PMC8106230 DOI: 10.1186/s40478-021-01190-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (mTBI) results in broad neurological symptoms and an increased risk of being diagnosed with a neurodegenerative disease later in life. While the immediate oxidative stress response and post-mortem pathology of the injured brain has been well studied, it remains unclear how early pathogenic changes may drive persistent symptoms and confer susceptibility to neurodegeneration. In this study we have used a mouse model of repeated mTBI (rmTBI) to identify early gene expression changes at 24 h or 7 days post-injury (7 dpi). At 24 h post-injury, gene expression of rmTBI mice shows activation of the DNA damage response (DDR) towards double strand DNA breaks, altered calcium and cell–cell signalling, and inhibition of cell death pathways. By 7 dpi, rmTBI mice had a gene expression signature consistent with induction of cellular senescence, activation of neurodegenerative processes, and inhibition of the DDR. At both timepoints gliosis, microgliosis, and axonal damage were evident in the absence of any gross lesion, and by 7 dpi rmTBI also mice had elevated levels of IL1β, p21, 53BP1, DNA2, and p53, supportive of DNA damage-induced cellular senescence. These gene expression changes reflect establishment of processes usually linked to brain aging and suggests that cellular senescence occurs early and most likely prior to the accumulation of toxic proteins. These molecular changes were accompanied by spatial learning and memory deficits in the Morris water maze. To conclude, we have identified DNA damage-induced cellular senescence as a repercussion of repeated mild traumatic brain injury which correlates with cognitive impairment. Pathways involved in senescence may represent viable treatment targets of post-concussive syndrome. Senescence has been proposed to promote neurodegeneration and appears as an effective target to prevent long-term complications of mTBI, such as chronic traumatic encephalopathy and other related neurodegenerative pathologies.
Collapse
|
44
|
Mechanical stimulation induced osteogenic differentiation of BMSCs through TWIST/E2A/p21 axis. Biosci Rep 2021; 40:222707. [PMID: 32309849 PMCID: PMC7199451 DOI: 10.1042/bsr20193876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/02/2020] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
The relationship between mechanical force and alveolar bone remodeling is an important issue in orthodontics because tooth movement is dependent on the response of bone tissue to the mechanical force induced by the appliances used. Mechanical cyclical stretch plays an essential role in the cell osteogenic differentiation involved in bone remodeling. However, the underlying mechanisms are unclear, particularly the molecular pathways regulated by mechanical stimulation. In the present study, we reported a dynamic change of p21 level in response to mechanical cyclical stretch, and shRNA-p21 in bone marrow mesenchymal stem cells (BMSCs) induced osteogenic differentiation. The mechanism was mediated through TWIST/E2A/p21 axis. These results supported the mechanical stimulation-induced osteogenic differentiation is negatively regulated by p21.
Collapse
|
45
|
Stavropoulou E, Kantartzi K, Tsigalou C, Aftzoglou K, Voidarou C, Konstantinidis T, Chifiriuc MC, Thodis E, Bezirtzoglou E. Microbiome, Immunosenescence, and Chronic Kidney Disease. Front Med (Lausanne) 2021; 8:661203. [PMID: 33816535 PMCID: PMC8017168 DOI: 10.3389/fmed.2021.661203] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/23/2021] [Indexed: 01/10/2023] Open
Abstract
The gut microbiome is known as an important predictive tool for perceiving characteristic shifts in disease states. Multiple renal diseases and pathologies seem to be associated with gut dysbiosis which directly affects host homeostasis. The gastrointestinal-kidney dialogue confers interesting information about the pathogenesis of multiple kidney diseases. Moreover, aging is followed by specific shifts in the human microbiome, and gradual elimination of physiological functions predisposes the microbiome to inflammaging, sarcopenia, and disease. Aging is characterized by a microbiota with an abundance of disease-associated pathobionts. Multiple factors such as the immune system, environment, medication, diet, and genetic endowment are involved in determining the age of the microbiome in health and disease. Our present review promotes recently acquired knowledge and is expected to inspire researchers to advance studies and investigations on the involved pathways of the gut microbiota and kidney axis.
Collapse
Affiliation(s)
- Elisavet Stavropoulou
- CHUV (Centre Hospitalier Universitaire Vaudois), Rue du Bugnon, Lausanne, Switzerland.,Department of Infectious Diseases, Central Institute, Valais Hospital, Sion, Switzerland
| | - Konstantia Kantartzi
- Nephrology Clinic, Department of Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Department of Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| | | | | | | | - Mariana Carmen Chifiriuc
- Laboratory of Microbiology, Faculty of Biology, The Research Institute of the University of Bucharest (ICUB), University of Bucharest, Bucharest, Romania
| | - Elias Thodis
- Nephrology Clinic, Department of Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupoli, Greece
| |
Collapse
|
46
|
Saoudaoui S, Bernard M, Cardin GB, Malaquin N, Christopoulos A, Rodier F. mTOR as a senescence manipulation target: A forked road. Adv Cancer Res 2021; 150:335-363. [PMID: 33858600 DOI: 10.1016/bs.acr.2021.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cellular senescence, cancer and aging are highly interconnected. Among many important molecular machines that lie at the intersection of this triad, the mechanistic (formerly mammalian) target of rapamycin (mTOR) is a central regulator of cell metabolism, proliferation, and survival. The mTOR signaling cascade is essential to maintain cellular homeostasis in normal biological processes or in response to stress, and its dysregulation is implicated in the progression of many disorders, including age-associated diseases. Accordingly, the pharmacological implications of mTOR inhibition using rapamycin or others rapalogs span the treatment of various human diseases from immune disorders to cancer. Importantly, rapamycin is one of the only known pan-species drugs that can extend lifespan. The molecular and cellular mechanisms explaining the phenotypic consequences of mTOR are vast and heavily studied. In this review, we will focus on the potential role of mTOR in the context of cellular senescence, a tumor suppressor mechanism and a pillar of aging. We will explore the link between senescence, autophagy and mTOR and discuss the opportunities to exploit senescence-associated mTOR functions to manipulate senescence phenotypes in age-associated diseases and cancer treatment.
Collapse
Affiliation(s)
- Sarah Saoudaoui
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Monique Bernard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Guillaume B Cardin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Nicolas Malaquin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada
| | - Apostolos Christopoulos
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Otolaryngology-Head and Neck Surgery Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada; Institut du cancer de Montréal, Montreal, QC, Canada; Université de Montréal, Département de radiologie, radio-oncologie et médicine nucléaire, Montreal, QC, Canada.
| |
Collapse
|
47
|
Colaianni G, Errede M, Sanesi L, Notarnicola A, Celi M, Zerlotin R, Storlino G, Pignataro P, Oranger A, Pesce V, Tarantino U, Moretti B, Grano M. Irisin Correlates Positively With BMD in a Cohort of Older Adult Patients and Downregulates the Senescent Marker p21 in Osteoblasts. J Bone Miner Res 2021; 36:305-314. [PMID: 33053231 DOI: 10.1002/jbmr.4192] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/23/2020] [Accepted: 10/03/2020] [Indexed: 12/25/2022]
Abstract
Irisin is a myokine produced by skeletal muscle during exercise in both mice and humans. We previously showed that irisin treatment ameliorates immobility-induced osteoporosis and muscular atrophy in mice. Data in humans showed a positive association between irisin and bone mineral density (BMD) in athletes and a population of healthy children. However, the role of this myokine regarding the state of muscle and bone in the same population remained to be determined. For this purpose, 62 patients (age 68.71 ± 12.31 years) undergoing total hip or knee replacement were recruited. Our results showed that irisin serum levels negatively correlated with age (R = -0.515; p = .000018) and positively correlated with femoral BMD (R = 0.619; p = .001) and vertebral BMD (R = 0.201; p = .0001). Irisin was also positively associated with Fndc5 mRNA in muscle biopsies (R = 0.248; p = .016), as well as with Osteocalcin (Ocn) mRNA in bone biopsies (R = 0.708; p = .006). In skeletal muscle, FNDC5 positive fibers positively correlate with BMD of total femur (R = 0.765; p = .0014) and BMD of femoral neck (R = 0.575; p = .031), Interestingly, by analyzing patients divided by their T-score, we found lower irisin levels (p = .0011) in patients with osteopenia/osteoporosis (OP) compared to healthy controls matched for age and sex. By analyzing the senescence marker p21, we found a significant increase of its mRNA expression in the bone biopsies of OP patients compared to control ones. Therefore, we investigated in vitro whether rec-irisin had a direct effect on this senescence marker, showing that p21 mRNA expression was significantly downregulated in osteoblasts by the treatment with irisin. Overall, these results indicate that higher irisin levels are associated with a lower rate of age-related osteoporosis and that irisin could be effective in delaying the osteoblast aging process, suggesting a potential senolytic action of this myokine. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Angela Notarnicola
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Monica Celi
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Roberta Zerlotin
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Patrizia Pignataro
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Angela Oranger
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Vito Pesce
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Umberto Tarantino
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Biagio Moretti
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| |
Collapse
|
48
|
de la Torre P, Fernández-de la Torre M, Flores AI. Premature senescence of placental decidua cells as a possible cause of miscarriage produced by mycophenolic acid. J Biomed Sci 2021; 28:3. [PMID: 33397374 PMCID: PMC7780668 DOI: 10.1186/s12929-020-00704-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/23/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Successful pregnancy is supported by a healthy maternal-fetal interface (i.e., the decidual tissues) which holds the conceptus and safeguards it against stressors from the beginning of pregnancy. Any disturbance of this interface can presumably lead to the loss of pregnancy. The use of the immunosuppressive drug mycophenolic acid (MPA) should be discontinued in pregnancy given its abortive and embryotoxic effects. Direct teratogenic effects have been observed in mammalian embryos cultured in MPA, but the underlying mechanisms of abortion by MPA are less understood. METHODS Decidual stromal cells isolated from human placentas are cultured in the presence of clinically relevant doses of MPA. Data regarding the effects of MPA on the proliferation and viability of decidua cultures are first analysed and then, molecular pathways contributing to these effects are unravelled. RESULTS MPA treatment of decidual stromal cells results in loss of proliferation capacity and a decrease in the viability of decidua cultures. The molecular pathways involved in the effects of MPA on decidual stromal cells are a reduction in pre-rRNA synthesis and subsequent disruption of the nucleolus. The nucleolar stress stabilizes p53, which in turn, leads to a p21-mediated cell cycle arrest in late S and G2 phases, preventing the progression of the decidua cells into the mitosis. Furthermore, MPA does not induce apoptosis but activate mechanisms of autophagy and senescence in decidual stromal cells. CONCLUSION The irreversible growth arrest of decidua cells, whose role in the maintenance of the pregnancy microenvironment is known, may be one cause of miscarriage in MPA treated pregnant women.
Collapse
Affiliation(s)
- Paz de la Torre
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, Madrid, Spain
| | - Miguel Fernández-de la Torre
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, Madrid, Spain
| | - Ana I Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, Madrid, Spain.
| |
Collapse
|
49
|
DNA damage response and breast cancer development: Possible therapeutic applications of ATR, ATM, PARP, BRCA1 inhibition. DNA Repair (Amst) 2020; 98:103032. [PMID: 33494010 DOI: 10.1016/j.dnarep.2020.103032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most common and significant cancers in females regarding the loss of life quality. Similar to other cancers, one of the etiologic factors in breast cancer is DNA damage. A plethora of molecules are responsible for sensing DNA damage and mediating actions which lead to DNA repair, senescence, cell cycle arrest and if damage is unbearable to apoptosis. In each of these, aberrations leading to unrepaired damage was resulted in uncontrolled proliferation and cancer. Another cellular function is autophagy defined as a process eliminating of unnecessary proteins in stress cases involved in pathogenesis of cancer. Knowing their role in cancer, scholars have tried to develop strategies in order to target DDR and autophagy. Further, the interactions of DDR and autophagy plus their regulatory role on each other have been focused simultaneously. The present review study has aimed to illustrate the importance of DDR and autophagy in breast cancer according to the related studies and uncover the relation between DDR and autophagy and its significance in breast cancer therapy.
Collapse
|
50
|
Hendrychová D, Jorda R, Kryštof V. How selective are clinical CDK4/6 inhibitors? Med Res Rev 2020; 41:1578-1598. [PMID: 33300617 DOI: 10.1002/med.21769] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/28/2020] [Accepted: 11/29/2020] [Indexed: 12/29/2022]
Abstract
Pharmacological inhibition of cyclin-dependent kinase 4/6 (CDK4/6) has emerged as an efficient approach for treating breast cancer, and its clinical potential is expanding to other cancers. CDK4/6 inhibitors were originally believed to act by arresting proliferation in the G1 phase, but it is gradually becoming clear that the cellular response to these compounds is far more complex than this. Multiple context-dependent mechanisms of action are emerging, involving modulation of quiescence, senescence, autophagy, cellular metabolism, and enhanced tumor cell immunogenicity. These mechanisms may be driven by interactions with unexpected targets. We review cellular responses to the Food and Drug Administration-approved CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib, and summarize available knowledge of other drugs undergoing clinical trials, including data on their off-target landscapes. We emphasize the importance of comprehensively characterizing drugs' selectivity profiles to maximize their clinical efficacy and safety and to facilitate their repurposing to treat additional diseases based on their target spectrum.
Collapse
Affiliation(s)
- Denisa Hendrychová
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Radek Jorda
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Vladimír Kryštof
- Department of Experimental Biology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| |
Collapse
|