1
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [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: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
El-Hazek RMM, Zaher NH, Emam HES, El-Gazzar MG, Khalil A. Pyrazole-sulfonamide scaffold featuring dual-tail strategy as apoptosis inducers in colon cancer. Sci Rep 2023; 13:5782. [PMID: 37031294 PMCID: PMC10082777 DOI: 10.1038/s41598-023-32820-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/03/2023] [Indexed: 04/10/2023] Open
Abstract
Dual-tail strategy has been successfully utilized in the development of novel carbonic anhydrase IX (CA IX) inhibitors. Herein we adopted this approach in the design and synthesis of a series of novel pyridine sulfonamide-pyrazole hybrid scaffold mimicking dual-tail inhibitors of CA IX. A library of 15 compounds was synthesized and assessed for their potential cytotoxic effects against colorectal cancer cells. Compounds 3, and 11 induced potential cytotoxic effects against the three cancer cell lines (HCT-116, HT-29, and SW-620) with IC50s' of 45.88, 28.27, and 16.57 uM, 25.01, 8.99, and 3.27 µM, respectively. Both compounds induced cellular apoptosis on HCT-116 and SW-620 cells, while compound 3 induced necrosis as well. In addition, both compounds induced cell cycle arrest on G0/G1, and S phases. Also, compound 11 showed potential autophagy induction on both colon cancer cell lines (HCT-116, and HT-29), and a little bit on metastatic type. Both compounds were less cytotoxic than the reference drug on normal epithelial cell. The migration rates of HCT-116 and the metastatic one SW-620 were reduced by both compounds. Finally, molecular docking of compounds 3 and 11 into the active site of CA IX confirmed in vitro inhibitory activity for both compounds.
Collapse
Affiliation(s)
- Reham M M El-Hazek
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Nashwa H Zaher
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Hagar E S Emam
- Biomedical Research Division, Nawah Scientific, Cairo, Egypt
| | - Marwa G El-Gazzar
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Amira Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt.
| |
Collapse
|
3
|
Wu YZ, Su YH, Kuo CY. Stressing the Regulatory Role of Long Non-Coding RNA in the Cellular Stress Response during Cancer Progression and Therapy. Biomedicines 2022; 10:biomedicines10051212. [PMID: 35625948 PMCID: PMC9138696 DOI: 10.3390/biomedicines10051212] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/13/2022] Open
Abstract
Cellular stress response is an important adaptive mechanism for regulating cell fate decision when cells confront with stress. During tumorigenesis, tumor progression and the course of treatment, cellular stress signaling can activate subsequent response to deal with stress. Therefore, cellular stress response has impacts on the fate of tumor cells and tumor responsiveness relative to therapeutic agents. In recent years, attention has been drawn to long non-coding RNAs (lncRNAs), a novel class of RNA molecules with more than 200 nucleotides in length, which has little protein-coding potential and possesses various functions in multiple biological processes. Accumulating evidence has shown that lncRNAs are also engaged in the regulation of cellular stress response, particularly in cancers. Here, we summarize lncRNAs that have been reported in the adaptive response to major types of cellular stress including genotoxic, hypoxic, oxidative, metabolic and endoplasmic reticulum stress, all of which are often encountered by cancer cells. Specifically, the molecular mechanisms of how lncRNAs regulate cellular stress response during tumor progression or the development of therapy resistance are emphasized. The potential clinical applications of stress-responsive lncRNAs as biomarkers will also be discussed.
Collapse
Affiliation(s)
- Yi-Zhen Wu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan; (Y.-Z.W.); (Y.-H.S.)
| | - Yong-Han Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan; (Y.-Z.W.); (Y.-H.S.)
| | - Ching-Ying Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan; (Y.-Z.W.); (Y.-H.S.)
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 66909)
| |
Collapse
|
4
|
Kumari R, Ray AG, Mukherjee D, Chander V, Kar D, Kumar US, Bharadwaj P.V.P. D, Banerjee SK, Konar A, Bandyopadhyay A. Downregulation of PTEN Promotes Autophagy via Concurrent Reduction in Apoptosis in Cardiac Hypertrophy in PPAR α−/− Mice. Front Cardiovasc Med 2022; 9:798639. [PMID: 35224041 PMCID: PMC8881053 DOI: 10.3389/fcvm.2022.798639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/14/2022] [Indexed: 01/05/2023] Open
Abstract
Cardiac hypertrophy is characterized by an increase in the size of the cardiomyocytes which is initially triggered as an adaptive response but ultimately becomes maladaptive with chronic exposure to different hypertrophic stimuli. Prolonged cardiac hypertrophy is often associated with mitochondrial dysfunctions and cardiomyocyte cell death. Peroxisome proliferator activated receptor alpha (PPAR α), which is critical for mitochondrial biogenesis and fatty acid oxidation, is down regulated in hypertrophied cardiomyocytes. Yet, the role of PPAR α in cardiomyocyte death is largely unknown. To assess the role of PPAR α in chronic hypertrophy, isoproterenol, a β-adrenergic receptor agonist was administered in PPAR α knock out (PPAR α−/−) mice for 2 weeks and hypertrophy associated changes in cardiac tissues were observed. Echocardiographic analysis ensured the development of cardiac hypertrophy and compromised hemodynamics in PPAR α−/− mice. Proteomic analysis using high resolution mass spectrometer identified about 1,200 proteins enriched in heart tissue. Proteins were classified according to biological pathway and molecular functions. We observed an unexpected down regulation of apoptotic markers, Annexin V and p53 in hypertrophied heart tissue. Further validation revealed a significant down regulation of apoptosis regulator, PTEN, along with other apoptosis markers like p53, Caspase 9 and c-PARP. The autophagy markers Atg3, Atg5, Atg7, p62, Beclin1 and LC3 A/B were up regulated in PPAR α−/− mice indicating an increase in autophagy. Similar observations were made in a high cholesterol diet fed PPAR α−/−mice. The results were further validated in vitro using NRVMs and H9C2 cell line by blocking PPAR α that resulted in enhanced autophagosome formation upon hypertrophic stimulation. The results demonstrate that in the absence of PPAR α apoptotic pathway is inhibited while autophagy is enhanced. The data suggest that PPAR α signaling might act as a molecular switch between apoptosis and autophagy thereby playing a critical role in adaptive process in cardiac hypertrophy.
Collapse
Affiliation(s)
- Ritu Kumari
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Aleepta Guha Ray
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dibyanti Mukherjee
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Vivek Chander
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dipak Kar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Uppulapu Shravan Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Deepak Bharadwaj P.V.P.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Sanjay K. Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Aditya Konar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Arun Bandyopadhyay
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- *Correspondence: Arun Bandyopadhyay ; orcid.org/0000-0002-4885-7033
| |
Collapse
|
5
|
Feliz-Mosquea YR, Christensen AA, Wilson AS, Westwood B, Varagic J, Meléndez GC, Schwartz AL, Chen QR, Mathews Griner L, Guha R, Thomas CJ, Ferrer M, Merino MJ, Cook KL, Roberts DD, Soto-Pantoja DR. Combination of anthracyclines and anti-CD47 therapy inhibit invasive breast cancer growth while preventing cardiac toxicity by regulation of autophagy. Breast Cancer Res Treat 2018; 172:69-82. [PMID: 30056566 PMCID: PMC6195817 DOI: 10.1007/s10549-018-4884-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/10/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND A perennial challenge in systemic cytotoxic cancer therapy is to eradicate primary tumors and metastatic disease while sparing normal tissue from off-target effects of chemotherapy. Anthracyclines such as doxorubicin are effective chemotherapeutic agents for which dosing is limited by development of cardiotoxicity. Our published evidence shows that targeting CD47 enhances radiation-induced growth delay of tumors while remarkably protecting soft tissues. The protection of cell viability observed with CD47 is mediated autonomously by activation of protective autophagy. However, whether CD47 protects cancer cells from cytotoxic chemotherapy is unknown. METHODS We tested the effect of CD47 blockade on cancer cell survival using a 2-dimensional high-throughput cell proliferation assay in 4T1 breast cancer cell lines. To evaluate blockade of CD47 in combination with chemotherapy in vivo, we employed the 4T1 breast cancer model and examined tumor and cardiac tissue viability as well as autophagic flux. RESULTS Our high-throughput screen revealed that blockade of CD47 does not interfere with the cytotoxic activity of anthracyclines against 4T1 breast cancer cells. Targeting CD47 enhanced the effect of doxorubicin chemotherapy in vivo by reducing tumor growth and metastatic spread by activation of an anti-tumor innate immune response. Moreover, systemic suppression of CD47 protected cardiac tissue viability and function in mice treated with doxorubicin. CONCLUSIONS Our experiments indicate that the protective effects observed with CD47 blockade are mediated through upregulation of autophagic flux. However, the absence of CD47 in did not elicit a protective effect in cancer cells, but it enhanced macrophage-mediated cancer cell cytolysis. Therefore, the differential responses observed with CD47 blockade are due to autonomous activation of protective autophagy in normal tissue and enhancement immune cytotoxicity against cancer cells.
Collapse
Affiliation(s)
- Yismeilin R Feliz-Mosquea
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Ashley A Christensen
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Brian Westwood
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
| | - Jasmina Varagic
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Giselle C Meléndez
- Internal Medicine, Section on Cardiovascular Medicine, Pathology Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Anthony L Schwartz
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qing-Rong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lesley Mathews Griner
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rajarshi Guha
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Craig J Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maria J Merino
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA
- Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David R Soto-Pantoja
- Department of Surgery, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC, 27157, USA.
- Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
6
|
Gemcitabine-Induced Autophagy Protects Human Lung Cancer Cells from Apoptotic Death. Lung 2016; 194:959-966. [PMID: 27604425 DOI: 10.1007/s00408-016-9936-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/02/2016] [Indexed: 01/01/2023]
Abstract
PURPOSE Gemcitabine has been used as a therapeutic drug combined with cisplatin for the treatment of lung cancer patients. However, the prognosis is poor due to acquired resistance. Accumulating studies have revealed that autophagy may contribute to the drug resistance. Therefore, the present study is aimed to clarify the mechanisms underlying gemcitabine-acquired resistance. METHODS SPC-A1 and A549 cells were incubated with gemcitabine followed by assessment of cell viability with MTT assays. GFP-LC3 transient transfection, MDC staining, and transmission electron microscopy were used to detect the change of autophagy at morphological level. Flow cytometry was used to monitor the effect of 3-MA on gemcitabine-induced apoptosis. Western blot analysis was used to detect the expression of p62, LC3, Beclin-1, ATG5, activated caspase 3, Bax, BNIP3, BNIP3L, and Bcl-2. RESULTS Our study showed that gemcitabine significantly induced both autophagy and apoptosis in human lung cancer cells SPC-A1 and A549. Of interest was that when autophagy was inhibited by 3-MA, the gemcitabine-induced apoptosis was effectively enhanced, suggesting that gemcitabine can activate autophagy to impair the chemosensitivity of lung cancer cells. Furthermore, the inhibition of autophagy by 3-MA further increased the expression of activated caspase 3, Bax, BNIP3, and BNIP3L, all are critical apoptotic mediators. Contrarily, 3-MA treatment further decreased the expression of Bcl-2, which is an important anti-apoptotic protein. CONCLUSION Our study indicated that autophagy protected human lung cancer cells from gemcitabine-induced apoptosis, and the combined use of gemcitabine and an autophagic inhibitor in lung cancer patients may be an effective therapeutic strategy.
Collapse
|
7
|
Wu HM, Jiang ZF, Ding PS, Shao LJ, Liu RY. Hypoxia-induced autophagy mediates cisplatin resistance in lung cancer cells. Sci Rep 2015. [PMID: 26201611 PMCID: PMC4511870 DOI: 10.1038/srep12291] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia which commonly exists in solid tumors, leads to cancer cells chemoresistance via provoking adaptive responses including autophagy. Therefore, we sought to evaluate the role of autophagy and hypoxia as well as the underlying mechanism in the cisplatin resistance of lung cancer cells. Our study demonstrated that hypoxia significantly protected A549 and SPC-A1 cells from cisplatin-induced cell death in a Hif-1α- and Hif-2α- dependent manner. Moreover, compared with normoxia, cisplatin-induced apoptosis under hypoxia was markedly reduced. However, when autophagy was inhibited by 3-MA or siRNA targeted ATG5, this reduction was effectively attenuated, which means autophagy mediates cisplatin resisitance under hypoxia. In parallel, we showed that hypoxia robustly augmented cisplatin-induced autophagy activation, accompanying by suppressing cisplatin-induced BNIP3 death pathways, which was due to the more efficient autophagic process under hypoxia. Consequently, we proposed that autophagy was a protective mechanism after cisplatin incubation under both normoxia and hypoxia. However, under normoxia, autophagy activation ‘was unable to counteract the stress induced by cisplatin, therefore resulting in cell death, whereas under hypoxia, autophagy induction was augmented that solved the cisplatin-induced stress, allowing the cells to survival. In conclusion, augmented induction of autophagy by hypoxia decreased lung cancer cells susceptibility to cisplatin-induced apoptosis.
Collapse
Affiliation(s)
- Hui-Mei Wu
- Department of Pulmonary, Anhui Geriatric Institute, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Zi-Feng Jiang
- Department of Pulmonary, Anhui Geriatric Institute, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Pei-Shan Ding
- Department of Pulmonary, Anhui Geriatric Institute, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Li-Jie Shao
- Department of Pulmonary, Anhui Geriatric Institute, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| | - Rong-Yu Liu
- Department of Pulmonary, Anhui Geriatric Institute, the First Affiliated Hospital of Anhui Medical University, Jixi Road 218, Hefei 230022, China
| |
Collapse
|
8
|
Lao Y, Wan G, Liu Z, Wang X, Ruan P, Xu W, Xu D, Xie W, Zhang Y, Xu H, Xu N. The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 2014; 10:736-49. [PMID: 24642486 PMCID: PMC5119057 DOI: 10.4161/auto.28034] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Metabolic stress induces autophagy as an alternative source of energy and metabolites. Insufficient autophagy in nutrient-deprived cancer cells would be beneficial for cancer therapy. Here, we performed a functional screen in search of novel autophagy regulators from natural products. We showed that oblongifolin C (OC), a natural small molecule compound extracted from Garcinia yunnanensis Hu, is a potent autophagic flux inhibitor. Exposure to OC results in an increased number of autophagosomes and impaired degradation of SQSTM1/p62. Costaining of GFP-LC3B with LysoTracker Red or LAMP1 antibody demonstrates that autophagosome-lysosome fusion is blocked by OC treatment. Furthermore, OC inhibits lysosomal proteolytic activity by altering lysosomal acidification and downregulating the expression of lysosomal cathepsins. Importantly, OC can eliminate the tolerance of cancer cells to nutrient starvation. Starvation dramatically increases the susceptibility of cancer cells to OC-induced CASP3-dependent apoptosis in vitro. Subsequent studies in xenograft mouse model showed that OC has anticancer potency as revealed by increased staining of cleaved CASP3, LC3 puncta, and SQSTM1, as well as reduced expression of lysosomal cathepsins. Combined treatment with OC and caloric restriction potentiates anticancer efficacy of OC in vivo. Collectively, these data demonstrated that OC is a novel autophagic flux inhibitor and might be useful in anticancer therapy.
Collapse
Affiliation(s)
- Yuanzhi Lao
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Gang Wan
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Zhenyan Liu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Xiaoyu Wang
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Ping Ruan
- Department of Pathology; Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine; Nanning, China
| | - Wei Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China; School of Life Sciences; Tsinghua University; Beijing, China
| | - Danqing Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China; Department of Discovery Technologies; Roche R&D Center (China) Ltd; Shanghai, China
| | - Weidong Xie
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Yaou Zhang
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| | - Hongxi Xu
- School of Pharmacy; Shanghai University of Traditional Chinese Medicine; Shanghai, China
| | - Naihan Xu
- Key Lab in Healthy Science and Technology; Division of Life Science; Graduate School at Shenzhen; Tsinghua University; Shenzhen,China
| |
Collapse
|
9
|
The miR-290-295 cluster suppresses autophagic cell death of melanoma cells. Sci Rep 2012; 2:808. [PMID: 23150779 PMCID: PMC3496171 DOI: 10.1038/srep00808] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/17/2012] [Indexed: 12/04/2022] Open
Abstract
We compared the expression levels of 307 miRNAs in six different B16F1 melanoma cell lines of differing malignant properties and found that the miR-290–295 cluster showed a strong upregulation in the more malignant B16F1 daughter cell lines. Its overexpression in B16F1 cells had no major effects on cell proliferation, migration or anchorage-independent growth, but conferred resistance to glucose starvation. This was mediated by miR-290-295-induced downregulation of several essential autophagy genes, including Atg7 and ULK1, which resulted in inhibition of autophagic cell death induced by glucose starvation. Similar effects were observed after knockdown of Atg7 or ULK1 in B16F1 melanoma cells, and after treatment with two chemical inhibitors of autophagy. Together, these results indicate that autophagy mediates cell death of melanoma cells under chronic nutrient deprivation, and they reveal an unanticipated role of the miR-290-295 cluster in conferring a survival advantage to melanoma cells by inhibiting autophagic cell death.
Collapse
|
10
|
Fan R, Miao Y, Shan X, Qian H, Song C, Wu G, Chen Y, Zha W. Bif-1 is overexpressed in hepatocellular carcinoma and correlates with shortened patient survival. Oncol Lett 2012; 3:851-854. [PMID: 22741005 DOI: 10.3892/ol.2012.562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 12/21/2011] [Indexed: 11/06/2022] Open
Abstract
Bax-interacting factor-1 (Bif-1) interacts with Beclin1 [the mammalian ortholog of yeast autophagy-related gene 6 (Atg6)] and affects the formation of autophagosomes during autophagy. The aim of this study was to explore Bif-1 expression and its prognostic significance in comparison with various clinicopathological predictors of survival. Bif-1 protein expression was determined by immunohistochemistry in 206 hepatocellular carcinomas. Cytoplasmic immunoreactivity was scored semi-quantitatively. The results were analyzed in correlation with various clinicopathological characteristics, including patient survival. The Chi-square test and Kaplan-Meier survival analysis were applied. The expression of Bif-1 was significantly higher in the hepatocellular cancers than in the adjacent matched non-tumor tissues (51.5 vs. 33.0%, P<0.01). Increased expression of Bif-1 in hepatocellular carcinomas was significantly correlated with a low grade of differentiation and a shortened overall survival (P<0.05). No significant differences were found between the expression of Bif-1 and age, gender, tumor size, damage of capsule, expression of hepatitis B surface antigen (HBs-Ag) and portal venous invasion. Our data demonstrated that Bif-1 is frequently expressed in hepatocellular carcinoma. Overexpression of Bif-1 is a new independent prognostic marker, which is associated with poor differentiation as well as shortened overall survival.
Collapse
Affiliation(s)
- Rengen Fan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Battisti S, Valente D, Albonici L, Bei R, Modesti A, Palumbo C. Nutritional stress and arginine auxotrophy confer high sensitivity to chloroquine toxicity in mesothelioma cells. Am J Respir Cell Mol Biol 2011; 46:498-506. [PMID: 22074703 DOI: 10.1165/rcmb.2011-0195oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The correlation between cell sensitivity to autophagy inhibitors, such as chloroquine (CQ), and the expression/activity of molecules involved in the control and execution of autophagy is well documented. However, tumor cells with comparable autophagic potentials may display variable degrees of autophagy addiction, due to the differential expression of molecular determinants, which are still scarcely defined. In this study, we investigated the effects of CQ on growth, death, and autophagic activity of malignant mesothelioma cell lines cultured in standard versus nutritional stress conditions partially mimicking those found in the tumor microenvironment. We report that, in each cell line, the toxic effects of CQ were amplified by nutritional stress and paralleled by autophagy inhibition. Still, the cell lines displayed different levels of sensitivity to CQ toxicity, which did not correlate with their relative degrees of constitutive and nutritional stress-induced autophagy, nor with the relative magnitude of the autophagy inhibition induced by the drug. Thus, we tested the hypothesis that the cell lines' sensitivity to CQ was related to their variable dependence on recycling of intracellular constituents by autophagy. In fact, the cell line with the highest sensitivity to the toxic effects of CQ was auxotrophic for arginine, due to the deficient expression of the enzyme argininosuccinate synthetase (ASS). Furthermore, overexpression of ASS in these cells reduced their sensitivity to CQ toxicity. Based on these results, the assessment of ASS expression in malignant mesothelioma tissues may allow the identification of subgroups of tumors with an increased sensitivity to the toxic effects of this drug.
Collapse
Affiliation(s)
- Sonia Battisti
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Italy
| | | | | | | | | | | |
Collapse
|
12
|
Zhang J, Wen HJ, Guo ZM, Zeng MS, Li MZ, Jiang YE, He XG, Sun CZ. TRB3 overexpression due to endoplasmic reticulum stress inhibits AKT kinase activation of tongue squamous cell carcinoma. Oral Oncol 2011; 47:934-9. [DOI: 10.1016/j.oraloncology.2011.06.512] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 06/19/2011] [Accepted: 06/25/2011] [Indexed: 11/29/2022]
|
13
|
Lacher MD, Pincheira RJ, Castro AF. Consequences of interrupted Rheb-to-AMPK feedback signaling in tuberous sclerosis complex and cancer. Small GTPases 2011; 2:211-216. [PMID: 22145093 DOI: 10.4161/sgtp.2.4.16703] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/02/2011] [Accepted: 06/06/2011] [Indexed: 02/06/2023] Open
Abstract
Rheb is a small GTPase primarily known for activating mammalian target of rapamycin complex 1 (mTORC1) and promoting cell growth in response to insulin and nutrients (amino acids, glucose). Shortage of glucose activates adenosine 5'-monophosphate-activated protein kinase (AMPK), which induces catabolic processes that produce ATP and suppresses energy-consuming anabolic reactions. As part of the latter response, AMPK activates the TSC1-TSC2 tumor suppressor complex, which in turn inhibits Rheb, thereby reducing mTORC1 activity and consequently suppressing protein synthesis. We recently identified an mTORC1-independent Rheb-to-AMPK feedback signaling loop in Tsc2-null in vitro models of Tuberous Sclerosis Complex (TSC). In addition to activating AMPK, Rheb reduced the nuclear levels of the cyclin-dependent kinase inhibitor p27(KIP1) (p27). Importantly, Rheb-mediated repression of p27 correlated with activation of Cdk2 and cell proliferation, and with tumor formation by TSC cells. Considering that AMPK was previously reported to regulate stability and subcellular localization of p27, we hypothesize that Rheb regulates p27 in TSC cells by activating AMPK. This article discusses how Rheb-to-AMPK, and p27 signaling may impact on disease progression and treatment of TSC, including sporadic lymphangioleiomyomatosis (S-LAM) and malignancies.
Collapse
Affiliation(s)
- Markus D Lacher
- Helen Diller Family Comprehensive Cancer Center; University of California; San Francisco, CA USA
| | | | | |
Collapse
|
14
|
Abstract
Macroautophagy (autophagy) is a lysosomal degradation pathway for the breakdown of intracellular proteins and organelles. Although constitutive autophagy is a homeostatic mechanism for intracellular recycling and metabolic regulation, autophagy is also stress responsive, in which it is important for the removal of damaged proteins and organelles. Autophagy thereby confers stress tolerance, limits damage, and sustains viability under adverse conditions. Autophagy is a tumor-suppression mechanism, yet it enables tumor cell survival in stress. Reconciling how loss of a prosurvival function can promote tumorigenesis, emerging evidence suggests that preservation of cellular fitness by autophagy may be key to tumor suppression. As autophagy is such a fundamental process, establishing how the functional status of autophagy influences tumorigenesis and treatment response is important. This is especially critical as many current cancer therapeutics activate autophagy. Therefore, efforts to understand and modulate the autophagy pathway will provide new approaches to cancer therapy and prevention.
Collapse
Affiliation(s)
- Eileen White
- The Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA.
| | | |
Collapse
|