1
|
Jana S, Alayash AI. Exploring the Molecular Interplay Between Oxygen Transport, Cellular Oxygen Sensing, and Mitochondrial Respiration. Antioxid Redox Signal 2025. [PMID: 39846399 DOI: 10.1089/ars.2023.0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Significance: The mitochondria play a key role in maintaining oxygen homeostasis under normal oxygen tension (normoxia) and during oxygen deprivation (hypoxia). This is a critical balancing act between the oxygen content of the blood, the tissue oxygen sensing mechanisms, and the mitochondria, which ultimately consume most oxygen for energy production. Recent Advances: We describe the well-defined role of the mitochondria in oxygen metabolism with a special focus on the impact on blood physiology and pathophysiology. Critical Issues: Fundamental questions remain regarding the impact of mitochondrial responses to changes in overall blood oxygen content under normoxic and hypoxic states and in the case of impaired oxygen sensing in various cardiovascular and pulmonary complications including blood disorders involving hemolysis and hemoglobin toxicity, ischemia reperfusion, and even in COVID-19 disease. Future Directions: Understanding the nature of the crosstalk among normal homeostatic pathways, oxygen carrying by hemoglobin, utilization of oxygen by the mitochondrial respiratory chain machinery, and oxygen sensing by hypoxia-inducible factor proteins, may provide a target for future therapeutic interventions. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| |
Collapse
|
2
|
Behera BP, Mishra SR, Patra S, Mahapatra KK, Bhol CS, Panigrahi DP, Praharaj PP, Klionsky DJ, Bhutia SK. Molecular regulation of mitophagy signaling in tumor microenvironment and its targeting for cancer therapy. Cytokine Growth Factor Rev 2025:S1359-6101(25)00004-8. [PMID: 39880721 DOI: 10.1016/j.cytogfr.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
Aberrations emerging in mitochondrial homeostasis are restrained by mitophagy to control mitochondrial integrity, bioenergetics signaling, metabolism, oxidative stress, and apoptosis. The mitophagy-accompanied mitochondrial processes that occur in a dysregulated condition act as drivers for cancer occurrence. In addition, the enigmatic nature of mitophagy in cancer cells modulates the cellular proteome, creating challenges for therapeutic interventions. Several reports found the role of cellular signaling pathways in cancer to modulate mitophagy to mitigate stress, immune checkpoints, energy demand, and cell death. Thus, targeting mitophagy to hinder oncogenic intracellular signaling by promoting apoptosis, in hindsight, might have an edge against cancer. This review highlights the receptors and adaptors, and the involvement of many proteins in mitophagy and their role in oncogenesis. It also provides insight into using mitophagy as a potential target for therapeutic intervention in various cancer types.
Collapse
Affiliation(s)
- Bishnu Prasad Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India.
| |
Collapse
|
3
|
Famta P, Shah S, Dey B, Kumar KC, Bagasariya D, Vambhurkar G, Pandey G, Sharma A, Srinivasarao DA, Kumar R, Guru SK, Raghuvanshi RS, Srivastava S. Despicable role of epithelial-mesenchymal transition in breast cancer metastasis: Exhibiting de novo restorative regimens. CANCER PATHOGENESIS AND THERAPY 2025; 3:30-47. [PMID: 39872366 PMCID: PMC11764040 DOI: 10.1016/j.cpt.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/30/2025]
Abstract
Breast cancer (BC) is the most prevalent cancer in women globally. Anti-cancer advancements have enabled the killing of BC cells through various therapies; however, cancer relapse is still a major limitation and decreases patient survival and quality of life. Epithelial-to-mesenchymal transition (EMT) is responsible for tumor relapse in several cancers. This highly regulated event causes phenotypic, genetic, and epigenetic changes in the tumor microenvironment (TME). This review summarizes the recent advancements regarding EMT using de-differentiation and partial EMT theories. We extensively review the mechanistic pathways, TME components, and various anti-cancer adjuvant and neo-adjuvant therapies responsible for triggering EMT in BC tumors. Information regarding essential clinical studies and trials is also discussed. Furthermore, we also highlight the recent strategies targeting various EMT pathways. This review provides a holistic picture of BC biology, molecular pathways, and recent advances in therapeutic strategies.
Collapse
Affiliation(s)
- Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Biswajit Dey
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Kondasingh Charan Kumar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Deepkumar Bagasariya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Dadi A. Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Rahul Kumar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | | | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
4
|
Yan T, Shi J. Angiogenesis and EMT regulators in the tumor microenvironment in lung cancer and immunotherapy. Front Immunol 2024; 15:1509195. [PMID: 39737184 PMCID: PMC11682976 DOI: 10.3389/fimmu.2024.1509195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Lung cancer remains the primary cause of cancer-related mortality, with factors such as postoperative tumor recurrence, metastasis, and therapeutic drug resistance exacerbating patient outcomes. Immunotherapy has emerged as a transformative approach, challenging conventional treatment paradigms for lung cancer. Consequently, advancing research in lung cancer immunotherapy is imperative. Recent studies indicate that numerous regulators within the tumor microenvironment (TME) drive tumor angiogenesis and epithelial-mesenchymal transition (EMT); these processes are interdependent, reciprocal, and collectively contribute to tumor progression. Tumor angiogenesis not only supplies adequate oxygen and nutrients for cellular proliferation but also establishes pathways facilitating tumor metastasis and creating hypoxic regions that foster drug resistance. Concurrently, EMT enhances metastatic potential and reinforces drug-resistance genes within tumor cells, creating a reciprocal relationship with angiogenesis. This interplay ultimately results in tumor invasion, metastasis, and therapeutic resistance. This paper reviews key regulators of angiogenesis and EMT, examining their impact on lung cancer immunotherapy and progression, and investigates whether newly identified regulators could influence lung cancer treatment, thus offering valuable insights for developing future therapeutic strategies.
Collapse
Affiliation(s)
- Taotao Yan
- Medical School of Nantong University, Nantong University, Nantong, China
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jiahai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
5
|
Wei M, Chen W, Dong Y, Gu Y, Wei D, Zhang J, Ren Y. Hypoxia-Inducible Factor-1α-Activated Protein Switch Based on Allosteric Self-Splicing Reduces Nonspecific Cytotoxicity of Pharmaceutical Drugs. Mol Pharm 2024; 21:5335-5347. [PMID: 39213620 DOI: 10.1021/acs.molpharmaceut.4c00921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Protein-based therapeutic agents currently used for targeted tumor therapy exhibit limited penetrability, nonspecific toxicity, and a short circulation half-life. Although targeting cell surface receptors improves cancer selectivity, the receptors are also slightly expressed in normal cells; consequently, the nonspecific toxicity of recombinant protein-based therapeutic agents has not been eliminated. In this study, an allosteric-regulated protein switch was designed that achieved cytoplasmic reorganization of engineered immunotoxins in tumor cells via interactions between allosteric self-splicing elements and cancer markers. It can target the accumulated HIF-1α in hypoxic cancer cells and undergo allosteric activation, and the splicing products were present in hypoxic cancer cells but were absent in normoxic cells, selectively killing tumor cells and reducing nonspecific toxicity to normal cells. The engineered pro-protein provides a platform for targeted therapy of tumors while offering a novel universal strategy for combining the activation of therapeutic functions with specific cancer markers. The allosteric self-splicing element is a powerful tool that significantly reduces the nonspecific cytotoxicity of therapeutic proteins.
Collapse
Affiliation(s)
- Min Wei
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Wenxin Chen
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuguo Dong
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yiyang Gu
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Dongzhi Wei
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jian Zhang
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yuhong Ren
- State Key Laboratory of Bioreactor Engineering, New World Institute of Biotechnology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
6
|
Al-Ostoot FH, Salah S, Khanum SA. An Overview of Cancer Biology, Pathophysiological Development and It's Treatment Modalities: Current Challenges of Cancer anti-Angiogenic Therapy. Cancer Invest 2024; 42:559-604. [PMID: 38874308 DOI: 10.1080/07357907.2024.2361295] [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: 02/17/2021] [Revised: 11/22/2021] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
A number of conditions and factors can cause the transformation of normal cells in the body into malignant tissue by changing the normal functions of a wide range of regulatory, apoptotic, and signal transduction pathways. Despite the current deficiency in fully understanding the mechanism of cancer action accurately and clearly, numerous genes and proteins that are causally involved in the initiation, progression, and metastasis of cancer have been identified. But due to the lack of space and the abundance of details on this complex topic, we have emphasized here more recent advances in our understanding of the principles implied tumor cell transformation, development, invasion, angiogenesis, and metastasis. Inhibition of angiogenesis is a significant strategy for the treatment of various solid tumors, that essentially depend on cutting or at least limiting the supply of blood to micro-regions of tumors, leading to pan-hypoxia and pan-necrosis inside solid tumor tissues. Researchers have continued to enhance the efficiency of anti-angiogenic drugs over the past two decades, to identify their potential in the drug interaction, and to discover reasonable interpretations for possible resistance to treatment. In this review, we have discussed an overview of cancer history and recent methods use in cancer therapy, focusing on anti-angiogenic inhibitors targeting angiogenesis formation. Further, this review has explained the molecular mechanism of action of these anti-angiogenic inhibitors in various tumor types and their limitations use. In addition, we described the synergistic mechanisms of immunotherapy and anti-angiogenic therapy and summarizes current clinical trials of these combinations. Many phase III trials found that combining immunotherapy and anti-angiogenic therapy improved survival. Therefore, targeting the source supply of cancer cells to grow and spread with new anti-angiogenic agents in combination with different conventional therapy is a novel method to reduce cancer progression. The aim of this paper is to overview the varying concepts of cancer focusing on mechanisms involved in tumor angiogenesis and provide an overview of the recent trends in anti-angiogenic strategies for cancer therapy.
Collapse
Affiliation(s)
- Fares Hezam Al-Ostoot
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
- Department of Biochemistry, Faculty of Education & Science, Albaydha University, Al-Baydha, Yemen
| | - Salma Salah
- Faculty of Medicine and Health Sciences, Thamar University, Dhamar, Yemen
| | - Shaukath Ara Khanum
- Department of Chemistry, Yuvaraja's College, University of Mysore, Mysuru, India
| |
Collapse
|
7
|
Masarkar N, Pal M, Roy M, Yadav AK, Pandya B, Lokhande S, Kanwar JR, Ray SK, Mukherjee S. In-silico screening of bioactive compounds of Moringa oleifera as potential inhibitors targeting HIF-1α/VEGF/GLUT-1 pathway against Breast Cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024; 0:jcim-2024-0176. [PMID: 39024644 DOI: 10.1515/jcim-2024-0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES Breast cancer is among the most heterogeneous and aggressive diseases and a foremost cause of death in women globally. Hypoxic activation of HIF-1α in breast cancers triggers the transcription of a battery of genes encoding proteins that facilitate tumor growth and metastasis and is correlated with a poor prognosis. Based on the reported cytotoxic and anti-cancer properties of Moringa oleifera (Mo), this study explores the inhibitory effect of bioactive compounds from M. oleifera and breast cancer target proteins HIF-1α, VEGF, and GLUT-1 in silico. METHODS The X-ray crystallographic structures of HIF-1α, VEGF, and GLUT1 were sourced from the Protein Data Bank (PDB) and docked with 70 3D PubChem structures of bioactive compounds of M. oleifera using AutoDock Vina, and binding modes were analyzed using Discovery Studio. Five compounds with the highest binding energies were selected and further drug-likeness, oral bioavailability, ADME, and toxicity profiles were analyzed using SwissADME, ADMETSaR, and ADMETlab 3.0 web server. RESULTS Out of the screened 70 bioactive compounds, the top five compounds with the best binding energies were identified namely Apigenin, Ellagic Acid, Isorhamnetin, Luteolin, and Myricetin with each receptor. Molecular docking results indicated that the ligands interact strongly with the target HIF-1α, VEGF, and GLUT-1 receptors through hydrogen bonds and hydrophobic interactions. These compounds showed favorable drug-like and pharmacokinetic properties, possessed no substantial toxicity, and were fairly bioavailable. CONCLUSIONS Results suggested that the compounds possess strong potential in developing putative lead compounds targeting HIF-1α that are safe natural plant-based drugs against breast cancer.
Collapse
Affiliation(s)
- Neha Masarkar
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Imphal, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Imphal, India
| | - Ashish K Yadav
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Bharati Pandya
- Department of General Surgery, All India Institute of Medical Sciences (AIIMS) Bhopal, Bhopal, India
| | - Suryabhan Lokhande
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | - Jagat R Kanwar
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| | | | - Sukhes Mukherjee
- Department of Biochemistry, 390706 All India Institute of Medical Sciences (AIIMS) Bhopal , Bhopal, Madhya Pradesh, India
| |
Collapse
|
8
|
Elsebaie HA, Nafie MS, Tawfik HO, Belal A, Ghoneim MM, Obaidullah AJ, Shaaban S, Ayed AA, El-Naggar M, Mehany ABM, Shaldam MA. Discovery of new 1,3-diphenylurea appended aryl pyridine derivatives as apoptosis inducers through c-MET and VEGFR-2 inhibition: design, synthesis, in vivo and in silico studies. RSC Med Chem 2024; 15:2553-2569. [PMID: 39026631 PMCID: PMC11253870 DOI: 10.1039/d4md00280f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Interest has been generated in VEGFR-2 and c-MET as potential receptors for the treatment of different malignancies. Using aryl pyridine derivatives with 1,3-diphenylurea attached, a number of promising dual VEGFR-2 and c-MET inhibitors were developed and synthesized. Regarding the molecular target, compounds 2d, 2f, 2j, 2k, and 2n had potent IC50 values of 65, 24, 150, 170, and 18 nM against c-MET, respectively. Additionally, they had potent IC50 values of 310, 35, 290, 320, and 24 nM against VEGFR-2, respectively. Regarding cytotoxicity, compounds 2d, 2f, 2j, 2k and 2n exhibited potent cytotoxicity against MCF-7 with IC50 values in the range 0.76-21.5 μM, and they showed promising cytotoxic activity against PC-3 with IC50 values in the range 1.85-3.42 μM compared to cabozantinib (IC50 = 1.06 μM against MCF-7 and 2.01 μM against PC-3). Regarding cell death, compound 2n caused cell death in MCF-7 cells by 87.34-fold; it induced total apoptosis by 33.19% (8.04% for late apoptosis, 25.15% for early apoptosis), stopping their growth in the G2/M phase, affecting the expression of apoptosis-related genes P53, Bax, caspases 3 and 9 and the anti-apoptotic gene, Bcl-2. In vivo study illustrated the anticancer activity of compound 2n by reduction of tumor mass and volume, and the tumor inhibition ratio reached 56.1% with an improvement of hematological parameters. Accordingly, compound 2n can be further developed as a selective target-oriented chemotherapeutic against breast cancer.
Collapse
Affiliation(s)
- Heba A Elsebaie
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University Tanta 31527 Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah Sharjah 27272 United Arab Emirates
- Chemistry Department, Faculty of Science, Suez Canal University Ismailia 41522 Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University Tanta 31527 Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AlSalam University in Egypt Kafr Al Zaiyat 6615062 Egypt
| | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University Ad Diriyah Riyadh 13713 Saudi Arabia
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University P.O. Box 2457 Riyadh 11451 Saudi Arabia
| | - Salwa Shaaban
- Department of Microbiology & Immunology, Faculty of pharmacySuef University Beni-Suef Egypt
- Department of Clinical Laboratory Sciences, Faculty of Applied medical Sciences, King Khalid University Abha Saudi Arabia
| | - Abdelmoneim A Ayed
- Department of Chemistry, Faculty of Science, Cairo University Giza Cairo 12613 Egypt
| | - Mohamed El-Naggar
- Chemistry department, Faculty of Sciences, Pure and Applied Chemistry Group, University of Sharjah P. O. Box 27272 Sharjah United Arab Emirates
| | - Ahmed B M Mehany
- Zoology Department, Faculty of Science (Boys), Al-Azhar University Cairo 11884 Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, AlSalam University in Egypt Kafr Al Zaiyat 6615062 Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University P.O. Box 33516 Kafrelsheikh Egypt
| |
Collapse
|
9
|
Fu J, Lin J, Dai Z, Lin B, Zhang J. Hypoxia-associated autophagy flux dysregulation in human cancers. Cancer Lett 2024; 590:216823. [PMID: 38521197 DOI: 10.1016/j.canlet.2024.216823] [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: 02/16/2024] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
A general feature of cancer is hypoxia, determined as low oxygen levels. Low oxygen levels may cause cells to alter in ways that contribute to tumor growth and resistance to treatment. Hypoxia leads to variations in cancer cell metabolism, angiogenesis and metastasis. Furthermore, a hypoxic tumor microenvironment might induce immunosuppression. Moreover, hypoxia has the potential to impact cellular processes, such as autophagy. Autophagy refers to the catabolic process by which damaged organelles and toxic macromolecules are broken down. The abnormal activation of autophagy has been extensively recorded in human tumors and it serves as a regulator of cell growth, spread to other parts of the body, and resistance to treatment. There is a correlation between hypoxia and autophagy in human malignancies. Hypoxia can regulate the activity of AMPK, mTOR, Beclin-1, and ATGs to govern autophagy in human malignancies. Furthermore, HIF-1α, serving as an indicator of low oxygen levels, controls the process of autophagy. Hypoxia-induced autophagy has a crucial role in regulating the growth, spread, and resistance to treatment in human malignancies. Hypoxia-induced regulation of autophagy can impact other mechanisms of cell death, such as apoptosis. Chemoresistance and radioresistance have become significant challenges in recent years. Hypoxia-mediated autophagy plays a crucial role in determining the response to these therapeutic treatments.
Collapse
Affiliation(s)
- Jiding Fu
- Department of Intensive Care Unit, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Baisheng Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China.
| |
Collapse
|
10
|
Jiang MQ, Yu SP, Estaba T, Choi E, Berglund K, Gu X, Wei L. Reprogramming Glioblastoma Cells into Non-Cancerous Neuronal Cells as a Novel Anti-Cancer Strategy. Cells 2024; 13:897. [PMID: 38891029 PMCID: PMC11171681 DOI: 10.3390/cells13110897] [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: 04/11/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Glioblastoma Multiforme (GBM) is an aggressive brain tumor with a high mortality rate. Direct reprogramming of glial cells to different cell lineages, such as induced neural stem cells (iNSCs) and induced neurons (iNeurons), provides genetic tools to manipulate a cell's fate as a potential therapy for neurological diseases. NeuroD1 (ND1) is a master transcriptional factor for neurogenesis and it promotes neuronal differentiation. In the present study, we tested the hypothesis that the expression of ND1 in GBM cells can force them to differentiate toward post-mitotic neurons and halt GBM tumor progression. In cultured human GBM cell lines, including LN229, U87, and U373 as temozolomide (TMZ)-sensitive and T98G as TMZ-resistant cells, the neuronal lineage conversion was induced by an adeno-associated virus (AAV) package carrying ND1. Twenty-one days after AAV-ND1 transduction, ND1-expressing cells displayed neuronal markers MAP2, TUJ1, and NeuN. The ND1-induced transdifferentiation was regulated by Wnt signaling and markedly enhanced under a hypoxic condition (2% O2 vs. 21% O2). ND1-expressing GBM cultures had fewer BrdU-positive proliferating cells compared to vector control cultures. Increased cell death was visualized by TUNEL staining, and reduced migrative activity was demonstrated in the wound-healing test after ND1 reprogramming in both TMZ-sensitive and -resistant GBM cells. In a striking contrast to cancer cells, converted cells expressed the anti-tumor gene p53. In an orthotopical GBM mouse model, AAV-ND1-reprogrammed U373 cells were transplanted into the fornix of the cyclosporine-immunocompromised C57BL/6 mouse brain. Compared to control GBM cell-formed tumors, cells from ND1-reprogrammed cultures formed smaller tumors and expressed neuronal markers such as TUJ1 in the brain. Thus, reprogramming using a single-factor ND1 overcame drug resistance, converting malignant cells of heterogeneous GBM cells to normal neuron-like cells in vitro and in vivo. These novel observations warrant further research using patient-derived GBM cells and patient-derived xenograft (PDX) models as a potentially effective treatment for a deadly brain cancer and likely other astrocytoma tumors.
Collapse
Affiliation(s)
- Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA 30033, USA
| | - Takira Estaba
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| | - Emily Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| |
Collapse
|
11
|
Dong F, Sun X, Su J, Li Q, He Y, Li W, Wang B, Wang B, Xu G, Wu X. Hypoxia-inducible PRMT2 addiction in glioblastomas. Cell Signal 2024; 117:111094. [PMID: 38341123 DOI: 10.1016/j.cellsig.2024.111094] [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: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Hypoxia-inducible transcription factors (HIFs) are key transcription factors for cellular response to low oxygen levels. However, the specific mediators responsible for activating downstream transcription are not well characterized. We previously identified Protein Arginine methyltransferase 2 (PRMT2), a highly expressed methyltransferase in glioblastoma multiforme, as a transcription co-activator. And we established a connection between PRMT2-mediated histone H3R8 asymmetric methylation (H3R8me2a) and transcription activation. Here we find that PRMT2 is activated by HIF1α under hypoxic conditions. And we demonstrate that PRMT2 and its H3R8me2a activity are required for the transcription activation of a significant subset of hypoxia-induced genes. Consequently, the inactivation of PRMT2 suppresses hypoxia-induced glioblastoma cell migration, attenuates tumor progression, and enhances chemotherapeutic sensitivity in mouse xenograft models. In addition, our analysis of clinical glioma specimens reveals a correlation between PRMT2 protein levels, HIF1α abundance, and an unfavorable prognosis. Our study establishes HIF1α-induced PRMT2 as a critical modulator in the activation of hypoxia-related transcriptional programs, ultimately driving malignant progression.
Collapse
Affiliation(s)
- Feng Dong
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, School of Biomedical Engineering & Technology, Tianjin Medical University, Tianjin 300070, China; Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Xiaoyu Sun
- Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Jiacheng Su
- Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Qian Li
- Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - You He
- Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
| | - Wei Li
- Department of Pathology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Baofeng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Bo Wang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative diseases, Tianjin Neurosurgical Institute, No. 6 Jizhao Road, Tianjin 300350, China
| | - Guogang Xu
- Health Management Institute, The Second Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China.
| | - Xudong Wu
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, School of Biomedical Engineering & Technology, Tianjin Medical University, Tianjin 300070, China; Department of Cell Biology, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China; Department of Neurosurgery, Tianjin Medical University General Hospital and Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin 300052, China.
| |
Collapse
|
12
|
Magar AG, Morya VK, Kwak MK, Oh JU, Noh KC. A Molecular Perspective on HIF-1α and Angiogenic Stimulator Networks and Their Role in Solid Tumors: An Update. Int J Mol Sci 2024; 25:3313. [PMID: 38542288 PMCID: PMC10970012 DOI: 10.3390/ijms25063313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 01/02/2025] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a major transcriptional factor, which plays an important role in cellular reprogramming processes under hypoxic conditions, which facilitate solid tumors' progression. HIF-1α is directly involved in the regulation of the angiogenesis, metabolic reprogramming, and extracellular matrix remodeling of the tumor microenvironment. Therefore, an in-depth study on the role of HIF-1α in solid tumor malignancies is required to develop novel anti-cancer therapeutics. HIF-1α also plays a critical role in regulating growth factors, such as the vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor, in a network manner. Additionally, it plays a significant role in tumor progression and chemotherapy resistance by regulating a variety of angiogenic factors, including angiopoietin 1 and angiopoietin 2, matrix metalloproteinase, and erythropoietin, along with energy pathways. Therefore, this review attempts to provide comprehensive insight into the role of HIF-1α in the energy and angiogenesis pathways of solid tumors.
Collapse
Affiliation(s)
- Anuja Gajanan Magar
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
- School of Medicine, Hallym University, Chuncheon-si 24252, Republic of Korea
| | - Vivek Kumar Morya
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Mi Kyung Kwak
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Ji Ung Oh
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| | - Kyu Cheol Noh
- Hallym University Dongtan Sacred Heart Hospital, Dongtan 18450, Republic of Korea
| |
Collapse
|
13
|
Moradi Hasan-Abad A, Shabankare A, Atapour A, Hamidi GA, Salami Zavareh M, Sobhani-Nasab A. The application of peroxidase mimetic nanozymes in cancer diagnosis and therapy. Front Pharmacol 2024; 15:1339580. [PMID: 38333005 PMCID: PMC10851941 DOI: 10.3389/fphar.2024.1339580] [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: 11/16/2023] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
In recent decades, scholarly investigations have predominantly centered on nanomaterials possessing enzyme-like characteristics, commonly referred to as nanozymes. These nanozymes have emerged as viable substitutes for natural enzymes, offering simplicity, stability, and superior performance across various applications. Inorganic nanoparticles have been extensively employed in the emulation of enzymatic activity found in natural systems. Nanoparticles have shown a strong ability to mimic a number of enzyme-like functions. These systems have made a lot of progress thanks to the huge growth in nanotechnology research and the unique properties of nanomaterials. Our presentation will center on the kinetics, processes, and applications of peroxidase-like nanozymes. In this discourse, we will explore the various characteristics that exert an influence on the catalytic activity of nanozymes, with a particular emphasis on the prevailing problems and prospective consequences. This paper presents a thorough examination of the latest advancements achieved in the domain of peroxidase mimetic nanozymes in the context of cancer diagnosis and treatment. The primary focus is on their use in catalytic cancer therapy, alongside chemotherapy, phototherapy, sonodynamic therapy, radiation, and immunotherapy. The primary objective of this work is to offer theoretical and technical assistance for the prospective advancement of anticancer medications based on nanozymes. Moreover, it is anticipated that this will foster the investigation of novel therapeutic strategies aimed at achieving efficacious tumor therapy.
Collapse
Affiliation(s)
- Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Atefe Shabankare
- Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholam Ali Hamidi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahmoud Salami Zavareh
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Sobhani-Nasab
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
14
|
Masarkar N, Ray SK, Saleem Z, Mukherjee S. Potential anti-cancer activity of Moringa oleifera derived bio-active compounds targeting hypoxia-inducible factor-1 alpha in breast cancer. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 0:jcim-2023-0182. [PMID: 37712721 DOI: 10.1515/jcim-2023-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/13/2023] [Indexed: 09/16/2023]
Abstract
Breast cancer (BC) will become a highly detected malignancy in females worldwide in 2023, with over 2 million new cases. Studies have established the role of hypoxia-inducible factor-1α (HIF1α), a transcription factor that controls cellular response to hypoxic stress, and is essential for BC spread. HIF-1 is implicated in nearly every critical stage of the metastatic progression, including invasion, EMT, intravasation, extravasation, angiogenesis, and the formation of metastatic niches. HIF-1 overexpression has been associated with poor prognosis and increased mortality in BC patients. This is accomplished by controlling the expression of HIF-1 target genes involved in cell survival, angiogenesis, metabolism, and treatment resistance. Studies have indicated that inhibiting HIF-1 has an anti-cancer effect on its own and that inhibiting HIF-1-mediated signaling improves the efficacy of anti-cancer therapy. Approximately 74 % of recognized anti-cancer drugs are sourced from plant species. Studies on anti-cancer characteristics of phytochemicals derived from Moringa oleifera (MO), also known as the 'Tree of Life', have revealed a high therapeutic potential for BC. In this review, we have highlighted the various mechanisms through which bioactive compounds present in MO may modulate HIF and its regulatory genes/pathways, to prove their efficacy in treating and preventing BC.
Collapse
Affiliation(s)
- Neha Masarkar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | | | - Zirha Saleem
- Department of Biotechnology, Institute for Excellence in Higher Education, Bhopal, Madhya Pradesh, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| |
Collapse
|
15
|
Dwivedi SK, Arachchige DL, Olowolagba A, Mahmoud M, Cunnien J, Tucker DR, Fritz D, Werner T, Luck RL, Liu H. Thiophene-based organic dye with large Stokes shift and deep red emission for live cell NAD(P)H detection under varying chemical stimuli. J Mater Chem B 2023; 11:6296-6307. [PMID: 37249441 PMCID: PMC10524713 DOI: 10.1039/d3tb00645j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
We report a novel method for synthesizing red and deep red cyanine dyes with large Stokes shifts, probes A and B, for live cell NAD(P)H detection. The probes were prepared using thiophene-based organic dyes featuring a π-conjugated bridge of thiophene and 3,4-ethylenedioxythiophene units linking the 1-methylquinolinium acceptor and formyl acceptor, respectively. These probes display weak absorption peaks at 315 nm (A) and 334 nm (B) and negligible fluorescence in the absence of NADH. However, upon the presence of NADH, new absorption and fluorescence peaks appear at 477 nm and 619 nm for probe A and at 486 nm and 576 nm for probe B, respectively. This is due to the NADH-facilitated reduction of the 1-methylquinolinium unit into 1-methyl-1,4-dihydroquinoline, which then acts as the electron donor for the probes, leading to the formation of well-defined electron donor-acceptor dye systems. Probe A has a large Stokes shift of 144 nm, which allows for better separation between the excitation and emission spectra, reducing spectral overlap and improving the accuracy of fluorescence measurements. The probes are highly selective for NAD(P)H, water-soluble, biocompatible, and easily permeable to cells. They are also photostable and were successfully used to monitor changes in NADH concentration in live cells during glycolysis in the presence of glucose, lactate, and pyruvate, treatment of FCCP and cancer drug cisplatin, and under hypoxia triggered by CoCl2. Furthermore, the probes were able to image NAD(P)H in Drosophila melanogaster larvae. Notably, cisplatin treatment increased the NAD(P)H concentration in A459 cells over time. Overall, this work presents a significant advancement in the field of live cell imaging by providing a simple and cost-effective method for detecting changes in NAD(P)H concentration under varying chemical stimuli.
Collapse
Affiliation(s)
- Sushil K Dwivedi
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Dilka Liyana Arachchige
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Adenike Olowolagba
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Mohamed Mahmoud
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Jenna Cunnien
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA
| | - Daniel R Tucker
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
| | - Delaney Fritz
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA
| | - Thomas Werner
- Department of Biological Sciences, Michigan Technological University, Houghton, MI 49931, USA
| | - Rudy L Luck
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
| | - Haiying Liu
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA.
| |
Collapse
|
16
|
Akimoto M, Susa T, Okudaira N, Koshikawa N, Hisaki H, Iizuka M, Okinaga H, Takenaga K, Okazaki T, Tamamori-Adachi M. Hypoxia induces downregulation of the tumor-suppressive sST2 in colorectal cancer cells via the HIF-nuclear IL-33-GATA3 pathway. Proc Natl Acad Sci U S A 2023; 120:e2218033120. [PMID: 37094129 PMCID: PMC10160999 DOI: 10.1073/pnas.2218033120] [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: 10/22/2022] [Accepted: 03/30/2023] [Indexed: 04/26/2023] Open
Abstract
As a decoy receptor, soluble ST2 (sST2) interferes with the function of the inflammatory cytokine interleukin (IL)-33. Decreased sST2 expression in colorectal cancer (CRC) cells promotes tumor growth via IL-33-mediated bioprocesses in the tumor microenvironment. In this study, we discovered that hypoxia reduced sST2 expression in CRC cells and explored the associated molecular mechanisms, including the expression of key regulators of ST2 gene transcription in hypoxic CRC cells. In addition, the effect of the recovery of sST2 expression in hypoxic tumor regions on malignant progression was investigated using mouse CRC cells engineered to express sST2 in response to hypoxia. Our results indicated that hypoxia-dependent increases in nuclear IL-33 interfered with the transactivation activity of GATA3 for ST2 gene transcription. Most importantly, hypoxia-responsive sST2 restoration in hypoxic tumor regions corrected the inflammatory microenvironment and suppressed tumor growth and lung metastasis. These results indicate that strategies targeting sST2 in hypoxic tumor regions could be effective for treating malignant CRC.
Collapse
Affiliation(s)
- Miho Akimoto
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Takao Susa
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Noriyuki Okudaira
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Nobuko Koshikawa
- Department of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Nitona, Chuoh-ku, Chiba260-8717, Japan
| | - Harumi Hisaki
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Masayoshi Iizuka
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
- Medical Education Center, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Hiroko Okinaga
- Department of Internal Medicine, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Keizo Takenaga
- Department of Innovative Cancer Therapeutics, Chiba Cancer Center Research Institute, Nitona, Chuoh-ku, Chiba260-8717, Japan
| | - Tomoki Okazaki
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| | - Mimi Tamamori-Adachi
- Department of Biochemistry, Teikyo University School of Medicine, Kaga, Itabashi-ku, Tokyo173-8605, Japan
| |
Collapse
|
17
|
Yang Y, Cheng C, He B, Du X, Liu J, Xia H, Wang P, Wu M, Wu H, Liu Q. Cigarette smoking, by accelerating the cell cycle, promotes the progression of non-small cell lung cancer through an HIF-1α-METTL3-m 6A/CDK2AP2 axis. JOURNAL OF HAZARDOUS MATERIALS 2023; 455:131556. [PMID: 37156046 DOI: 10.1016/j.jhazmat.2023.131556] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 04/09/2023] [Accepted: 05/01/2023] [Indexed: 05/10/2023]
Abstract
Cigarette smoking killed about 8 million people every year and promoted non-small cell lung cancer (NSCLC). We investigated the molecular mechanism of smoking-promoted NSCLC progression. Relative to non-smokers, NSCLC patients who were smokers had a higher tumor malignancy. For NSCLC cells, cigarette smoke extract (CSE) increased levels of HIF-1α, METTL3, Cyclin E1, and CDK2 and promoted the G1/S transition, which promoted cell proliferation. Down-regulation HIF-1α or METTL3 reversed these effects. meRIP-seq and RNA-seq revealed the m6A modification in Cyclin Dependent Kinase 2 Associated Protein 2 (CDK2AP2) mRNA as the key downstream target. Further, for NSCLC cells exposed to CSE, HIF-1α activated METTL3 transcription. Xenografts in nude mice demonstrated that HIF-1α via METTL3 participated in tumor growth. In NSCLC tissues of smokers, protein levels of HIF-1α and METTL3 were higher, and levels of CDK2AP2 were lower. In conclusion, HIF-1α via METTL3 regulation of the m6A modification of CDK2AP2 mRNA drives smoking-induced progression of NSCLC through promoting cell proliferation. This is a previously unknown molecular mechanism for smoking-induced NSCLC progression. The results have potential value for treatment of NSCLC, especially for patients who smoke.
Collapse
Affiliation(s)
- Yi Yang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Cheng Cheng
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Bin He
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Xuan Du
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Jinyuan Liu
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Haibo Xia
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Peiwen Wang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Meng Wu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Hao Wu
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Qizhan Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, China.
| |
Collapse
|
18
|
Lafleur VN, Halim S, Choudhry H, Ratcliffe PJ, Mole DR. Multi-level interaction between HIF and AHR transcriptional pathways in kidney carcinoma. Life Sci Alliance 2023; 6:e202201756. [PMID: 36725335 PMCID: PMC9896012 DOI: 10.26508/lsa.202201756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 02/03/2023] Open
Abstract
Hypoxia-inducible factor (HIF) and aryl hydrocarbon receptor (AHR) are members of the bHLH-PAS family of transcription factors that underpin cellular responses to oxygen and to endogenous and exogenous ligands, respectively, and have central roles in the pathogenesis of renal cancer. Composed of heterodimers, they share a common HIF-1β/ARNT subunit and similar DNA-binding motifs, raising the possibility of crosstalk between the two transcriptional pathways. Here, we identify both general and locus-specific mechanisms of interaction between HIF and AHR that act both antagonistically and cooperatively. Specifically, we observe competition for the common HIF-1β/ARNT subunit, in cis synergy for chromatin binding, and overlap in their transcriptional targets. Recently, both HIF and AHR inhibitors have been developed for the treatment of solid tumours. However, inhibition of one pathway may promote the oncogenic effects of the other. Therefore, our work raises important questions as to whether combination therapy targeting both of these pro-tumourigenic pathways might show greater efficacy than targeting each system independently.
Collapse
Affiliation(s)
| | - Silvia Halim
- NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Center of Innovation in Personalized Medicine, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter J Ratcliffe
- Ludwig Institute for Cancer Research, University of Oxford, Old Road Campus, Oxford, UK
- The Francis Crick Institute, London, UK
| | - David R Mole
- NDM Research Building, University of Oxford, Old Road Campus, Oxford, UK
| |
Collapse
|
19
|
Lan T, Ji N, Tian QQ, Zhan Y, He W. An edoplasmic reticulum-targeted NIR fluorescent probe with a large Stokes shift for hypoxia imaging. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 288:122201. [PMID: 36463622 DOI: 10.1016/j.saa.2022.122201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Hypoxia is closely linked to various diseases, including solid tumors. The level of nitroreductase (NTR) is usually abnormally upregulated in hypoxic conditions, which can be a biomarker of hypoxia. Herein, the first endoplasmic reticulum-targeting NIR fluorescent probe, ISO-NTR, was developed for highly selective and sensitive detection of NTR. It shows a large Stokes shift (185 nm) and a 5-fold increases in fluorescence intensity. Meanwhile, the ISO-NTR probe with a dicyanoisophorone derivative has excellent endoplasmic reticulum targeting in living systems with high Pearson's correlation coefficients (Rr = 0.9489). Molecular docking calculations and high binding energy between the probe and NTR (-10.78 kcal·mol-1) may explain the high selectivity of ISO-NTR. Additionally, it has been successfully applied to NTR imaging in vitro and vivo due to its good sensitivity, high selectivity and large Stokes shift, which may provide an effective method for studying the physiological and pathological functions of NTR in living systems. This probe could be developed as a potential imaging tool to further explore the pathogenesis of hypoxia-related diseases in endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ting Lan
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Nan Ji
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Qin-Qin Tian
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Yu Zhan
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China
| | - Wei He
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, PR China.
| |
Collapse
|
20
|
Ahmad MH, Ghosh B, Rizvi MA, Ali M, Kaur L, Mondal AC. Neural crest cells development and neuroblastoma progression: Role of Wnt signaling. J Cell Physiol 2023; 238:306-328. [PMID: 36502519 DOI: 10.1002/jcp.30931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/19/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is one of the most common heterogeneous extracranial cancers in infancy that arises from neural crest (NC) cells of the sympathetic nervous system. The Wnt signaling pathway, both canonical and noncanonical pathway, is a highly conserved signaling pathway that regulates the development and differentiation of the NC cells during embryogenesis. Reports suggest that aberrant activation of Wnt ligands/receptors in Wnt signaling pathways promote progression and relapse of NB. Wnt signaling pathways regulate NC induction and migration in a similar manner; it regulates proliferation and metastasis of NB. Inhibiting the Wnt signaling pathway or its ligands/receptors induces apoptosis and abrogates proliferation and tumorigenicity in all major types of NB cells. Here, we comprehensively discuss the Wnt signaling pathway and its mechanisms in regulating the development of NC and NB pathogenesis. This review highlights the implications of aberrant Wnt signaling in the context of etiology, progression, and relapse of NB. We have also described emerging strategies for Wnt-based therapies against the progression of NB that will provide new insights into the development of Wnt-based therapeutic strategies for NB.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Balaram Ghosh
- Department of Clinical Pharmacology, Midnapore Medical College & Hospital, West Bengal, Medinipur, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mansoor Ali
- School of Life Sciences, Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Loveleena Kaur
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine (IIIM), Srinagar, India
| | - Amal Chandra Mondal
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
21
|
Saleh MA, Antar SA, Abdo W, Ashour A, Zaki AA. Genistin modulates high-mobility group box protein 1 (HMGB1) and nuclear factor kappa-B (NF-κB) in Ehrlich-ascites-carcinoma-bearing mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:966-978. [PMID: 35907070 DOI: 10.1007/s11356-022-22268-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/23/2022] [Indexed: 06/15/2023]
Abstract
Cancer is the world's second-largest cause of death. Although there are numerous cancer treatment options, they are typically uncomfortable owing to side effects and ineffectual due to increased resistance to traditional anti-cancer medications or radiation therapy. A key method in cancer treatment is to target delayed/inhibited inflammation and apoptosis, which are very active areas of research. Natural chemicals originating from plants are of particular interest because of their high bioavailability, safety, few side effects, and, most importantly, cost-effectiveness. Flavonoids have become incredibly common as anti-cancer medications, with promising findings as cytotoxic anti-cancer agents that cause cancer cell death. Isolated compound (genistin) was evaluated for in vitro antiproliferative activity against breast cancer cell line (MCF-7 and MDA-MB-231). The compound exhibited good cytotoxic activities against both cell lines. In vivo antiproliferative efficacy was also investigated in Ehrlich's ascites carcinoma (EAC). Compared to the control group, genistin revealed a significant decrease in tumor weight, volume, high-mobility group box1 (HMGB1), and nuclear factor-kappa B (NF-κB) contents. On the other hand, B-cell lymphoma 2 (Bcl-2) contents increase suggesting an anti-inflammatory and anti-apoptotic activity through inhibition of HMGB1 signaling and activating the Bcl-2 pathway.
Collapse
Affiliation(s)
- Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| | - Walied Abdo
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516, Egypt
| | - Ahmed Ashour
- Department of Pharmacognosy, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed A Zaki
- Department of Pharmacognosy, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
22
|
Martins Pinto M, Paumard P, Bouchez C, Ransac S, Duvezin-Caubet S, Mazat JP, Rigoulet M, Devin A. The Warburg effect and mitochondrial oxidative phosphorylation: Friends or foes? BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2023; 1864:148931. [PMID: 36367492 DOI: 10.1016/j.bbabio.2022.148931] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Cancer cells display an altered energy metabolism, which was proposed to be the root of cancer. This early discovery was done by O. Warburg who conducted one of the first studies of tumor cell energy metabolism. Taking advantage of cancer cells that exhibited various growth rates, he showed that cancer cells display a decreased respiration and an increased glycolysis proportional to the increase in their growth rate, suggesting that they mainly depend on fermentative metabolism for ATP generation. Warburg's results and hypothesis generated controversies that are persistent to this day. It is thus of great importance to understand the mechanisms by which cancer cells can reversibly regulate the two pathways of their energy metabolism as well as the functioning of this metabolism in cell proliferation. In this review, we discuss of the origin of the decrease in cell respiratory rate, whether the Warburg effect is mandatory for an increased cell proliferation rate, the consequences of this effect on two major players of cell energy metabolism that are ATP and NADH, and the role of the microenvironment in the regulation of cellular respiration and metabolism both in cancer cell and in yeast.
Collapse
Affiliation(s)
- M Martins Pinto
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; CBMN, Allée de Geoffroy St Hilaire Bât, B1433600 Pessac, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - P Paumard
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - C Bouchez
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - S Ransac
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - S Duvezin-Caubet
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - J P Mazat
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - M Rigoulet
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France
| | - A Devin
- CNRS, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires, UMR 5095, F-33000 Bordeaux, France.
| |
Collapse
|
23
|
Shang C, Ou X, Zhang H, Wei D, Wang Q, Li G. Activation of PGRN/MAPK axis stimulated by the hypoxia-conditioned mesenchymal stem cell-derived HIF-1α facilitates osteosarcoma progression. Exp Cell Res 2022; 421:113373. [PMID: 36183781 DOI: 10.1016/j.yexcr.2022.113373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 09/13/2022] [Accepted: 09/25/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Progranulin (PGRN) is an important survival factor in the progression of multiple cancers. PURPOSE To explore the effects and mechanisms of PGRN on malignant biological behavior of osteosarcoma (OS) cells and the effects of mesenchymal stem cells (MSCs) and the hypoxic microenvironment on PGRN alteration. MATERIAL AND METHODS The expression pattern of PGRN in OS were evaluated in OS tissues and cell lines. Next, a loss-of-function assay investigated the function of PGRN on the proliferation, migration and cell death of OS cells. The activation of MAPK signaling in the process was examined by western blot and functional experiments accompanied by skatole. Additionally, we internally silenced hypoxia-inducible factor-1α (HIF-1α) in MSCs along with exogenously added HIF-1α (exo-HIF-1α) to explore how MSCs affect PGRN alteration and the malignant behavior of OS cells. RESULTS An aberrantly high expression of PGRN was observed in OS and associated with the poor prognosis of OS patients. PGRN knockdown repressed the proliferation, migration and induced cell death of OS cells, and activating MAPK pathway reversed these effects. Further evidence showed that MSCs regulated PGRN to mediate the malignant biological behavior of OS cells. Hypoxia enhanced HIF-1α expression in MSCs. HIF-1α silencing in MSCs under hypoxia suppressed the oncogenic effects of MSCs and reduced PGRN expression in OS cells, while the treatment of exo-HIF-1α reversed the depressive effects of HIF1α silencing on OS progression. CONCLUSION Overall, we concluded that PGRN, which was activated by the increase of hypoxic-MSCs-derived HIF-1α, promoted OS progression through the activation of MAPK signaling.
Collapse
Affiliation(s)
- Chi Shang
- Department of the Second Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Xuehai Ou
- Department of the First Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Hongxing Zhang
- Department of the Second Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Dengke Wei
- Department of the First Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Qiang Wang
- Department of the First Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China
| | - Gang Li
- Department of the First Hand Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710054, China.
| |
Collapse
|
24
|
Fernández-Palanca P, Payo-Serafín T, San-Miguel B, Méndez-Blanco C, Tuñón MJ, González-Gallego J, Mauriz JL. Hepatocellular carcinoma cells loss lenvatinib efficacy in vitro through autophagy and hypoxia response-derived neuropilin-1 degradation. Acta Pharmacol Sin 2022; 44:1066-1082. [PMID: 36376373 PMCID: PMC10104874 DOI: 10.1038/s41401-022-01021-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
AbstractDespite pharmacological advances such as lenvatinib approval, therapeutic failure of hepatocellular carcinoma (HCC) remains a big challenge due to the complexity of its underlying molecular mechanisms. Neuropilin-1 (NRP1) is a co-receptor involved in several cellular processes associated to chemoresistance development. Since both the double-edged process of autophagy and hypoxia-derived response play crucial roles in the loss of therapeutic effectiveness, herein we investigated the interplay among NRP1, autophagy and hypoxia in development of lenvatinib resistance in HCC cell lines. We first analyzed NRP1 expression levels in human HCC samples from public databases, found significantly increased NRP1 expression in human HCC samples as well as its correlation with advanced tumor and metastasis stages. Among 3 HCC cell lines (HepG2, Huh-7 and Hep3B), Hep3B and Huh-7 cells showed significantly increased NRP1 expression levels and cell migration ability together with higher susceptibility to lenvatinib. We demonstrated that NRP1 gene silencing significantly enhanced the anticancer effects of lenvatinib on Hep3B and Huh-7 cells. Furthermore, lenvatinib suppressed NRP1 expression through promoting autophagy in Hep3B and Huh-7 cells; co-treatment with bafilomycin A1 attenuated the antitumor effects of lenvatinib, and NRP1 silencing prevented this loss of in vitro effectiveness of lenvatinib even in the presence of bafilomycin A1. In addition, exposure to a hypoxic microenvironment significantly decreased NRP1 expression through autophagy in Hep3B and Huh-7 cells. Under hypoxia, HIF-1α directly modulated NRP1 expression; HIF-1α silencing not only enhanced the anticancer effects of combined lenvatinib and hypoxia, but also prevented the loss of effectiveness caused by bafilomycin A1, highlighting the potential role of HIF-1α-derived hypoxia response in the adaptive cellular response to lenvatinib and promoting resistance acquisition by autophagy modulation. Overall, NRP1 may constitute a potential therapeutic target to prevent lenvatinib failure derived from a hypoxia-associated modulation of autophagy in advanced HCC.
Collapse
|
25
|
Su Y, Li C, Liu W, Liu Y, Li L, Chen Q. Comprehensive analysis of differentially expressed miRNAs in mice with kidney injury induced by chronic intermittent hypoxia. Front Genet 2022; 13:918728. [DOI: 10.3389/fgene.2022.918728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022] Open
Abstract
Background: miRNAs have been reported to participate in various diseases. Nevertheless, the expression patterns of miRNA in obstructive sleep apnea (OSA)-induced kidney injury remain poorly characterized. In the current study, miRNA sequencing (miRNA-seq) was conducted to investigate miRNA expression profiles in a chronic intermittent hypoxia (CIH)-induced renal injury mouse model.Methods: The mouse model of chronic intermittent hypoxia was established. Differentially expressed miRNAs (DEmiRs) were detected using miRNA-seq technology. The sequencing data were subjected to Gene Ontology (GO) functional enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses using a bioinformatics approach. RT-qPCR was further used to evaluate the sequencing results. Finally, we created a network for clarifying the relationship between the miRNAs and target genes.Results: In total, nine miRNAs were identified to be upregulated and nine to be downregulated in a mouse model of renal injury induced by chronic intermittent hypoxia. The Kyoto Encyclopedia of Genes and Genomes analyses revealed that the Wnt signaling pathway was involved in the development of chronic intermittent hypoxia-induced renal injury. Subsequently, eight DEmiRs, namely, mmu-miR-486b–3p, mmu-miR-215–5p, mmu-miR-212–3p, mmu-miR-344–3p, mmu-miR-181b-1-3p, mmu-miR-467a–3p, mmu-miR-467 d-3p, and mmu-miR-96–5p, showed a similar trend of expression when verified using RT-qPCR. Finally, five selected DEmiRs were used to construct a miRNA–mRNA network.Conclusion: In conclusion, a total of 18 DEmiRs were identified in the mouse model of chronic intermittent hypoxia-induced renal injury. These findings advance our understanding of the molecular regulatory mechanisms underlying the pathophysiology of obstructive sleep apnea-associated chronic kidney disease.
Collapse
|
26
|
Ren W, Yuan Y, Peng J, Mutti L, Jiang X. The function and clinical implication of circular RNAs in lung cancer. Front Oncol 2022; 12:862602. [PMID: 36338714 PMCID: PMC9629004 DOI: 10.3389/fonc.2022.862602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 08/26/2022] [Indexed: 12/02/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Despite the recent advent of promising new targeted therapies, lung cancer diagnostic strategies still have difficulty in identifying the disease at an early stage. Therefore, the characterizations of more sensible and specific cancer biomarkers have become an important goal for clinicians. Circular RNAs are covalently close, endogenous RNAs without 5' end caps or 3'poly (A) tails and have been characterized by high stability, abundance, and conservation as well as display cell/tissue/developmental stage-specific expressions. Numerous studies have confirmed that circRNAs act as microRNA (miRNA) sponges, RNA-binding protein, and transcriptional regulators; some circRNAs even act as translation templates that participate in multiple pathophysiological processes. Growing evidence have confirmed that circRNAs are involved in the pathogenesis of lung cancers through the regulation of proliferation and invasion, cell cycle, autophagy, apoptosis, stemness, tumor microenvironment, and chemotherapy resistance. Moreover, circRNAs have emerged as potential biomarkers for lung cancer diagnosis and prognosis and targets for developing new treatments. In this review, we will summarize recent progresses in identifying the biogenesis, biological functions, potential mechanisms, and clinical applications of these molecules for lung cancer diagnosis, prognosis, and targeted therapy.
Collapse
Affiliation(s)
- Wenjun Ren
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yixiao Yuan
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Peng
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Luciano Mutti
- The Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Xiulin Jiang
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Bai R, Li Y, Jian L, Yang Y, Zhao L, Wei M. The hypoxia-driven crosstalk between tumor and tumor-associated macrophages: mechanisms and clinical treatment strategies. Mol Cancer 2022; 21:177. [PMID: 36071472 PMCID: PMC9454207 DOI: 10.1186/s12943-022-01645-2] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/25/2022] [Indexed: 02/08/2023] Open
Abstract
Given that hypoxia is a persistent physiological feature of many different solid tumors and a key driver for cancer malignancy, it is thought to be a major target in cancer treatment recently. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME), which have a large impact on tumor development and immunotherapy. TAMs massively accumulate within hypoxic tumor regions. TAMs and hypoxia represent a deadly combination because hypoxia has been suggested to induce a pro-tumorigenic macrophage phenotype. Hypoxia not only directly affects macrophage polarization, but it also has an indirect effect by altering the communication between tumor cells and macrophages. For example, hypoxia can influence the expression of chemokines and exosomes, both of which have profound impacts on the recipient cells. Recently, it has been demonstrated that the intricate interaction between cancer cells and TAMs in the hypoxic TME is relevant to poor prognosis and increased tumor malignancy. However, there are no comprehensive literature reviews on the molecular mechanisms underlying the hypoxia-mediated communication between tumor cells and TAMs. Therefore, this review has the aim to collect all recently available data on this topic and provide insights for developing novel therapeutic strategies for reducing the effects of hypoxia.
Collapse
Affiliation(s)
- Ruixue Bai
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.,Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China
| | - Lingyan Jian
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yuehui Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, People's Republic of China. .,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, People's Republic of China. .,Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, 110000, People's Republic of China.
| |
Collapse
|
28
|
Li W, Yuan P, Liu W, Xiao L, Xu C, Mo Q, Xu S, He Y, Jiang D, Wang X. Hypoxia–Immune-Related Gene SLC19A1 Serves as a Potential Biomarker for Prognosis in Multiple Myeloma. Front Immunol 2022; 13:843369. [PMID: 35958555 PMCID: PMC9358019 DOI: 10.3389/fimmu.2022.843369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Background Multiple myeloma (MM) remains an incurable malignant tumor of plasma cells. Increasing evidence has reported that hypoxia and immune status contribute to the progression of MM. In this research, the prognostic value of the hypoxia–immune-related gene SLC19A1 in MM was evaluated by bioinformatics analysis. Method RNA-sequencing (RNA-seq) data along with clinical information on MM were downloaded from the Gene Expression Omnibus (GEO) database. Consistent clustering analysis and ESTIMATE algorithms were performed to establish the MM sample subgroups related to hypoxia and immune status, respectively, based on the GSE24080 dataset. The differentially expressed analysis was performed to identify the hypoxia–immune-related genes. Subsequently, a hypoxia–immune-gene risk signature for MM patients was constructed by univariate and multivariate Cox regression analyses, which was also verified in the GSE4581 dataset. Furthermore, the mRNA expression of SLC19A1 was determined using qRT-PCR in 19 MM patients, and the correlations between the genetic expression of SLC19A1 and clinical features were further analyzed. Result A total of 47 genes were identified as hypoxia–immune-related genes for MM. Among these genes, SLC19A1 was screened to construct a risk score model that had better predictive power for MM. The constructed prognostic signature based on SLC19A1 was verified in the GSE4581 dataset. All independent prognostic factors (age, β2-microglobulin, LDH, albumin, MRI, and gene risk score) were used to develop a nomogram that showed a better performance for predicting the survival probability of MM patients for 1–5 years. Furthermore, SLC19A1 was highly expressed in newly diagnosed and relapsed MM patients, and high expression of SLC19A1 is correlated with higher bone marrow aspiration plasma cells and β2-microglobulin levels in MM patients. Conclusion In conclusion, our results suggest that SLC19A1 is aberrantly expressed in MM and highly expressed SLC19A1 might be a biomarker correlated with inferior prognosis. More importantly, we identified SLC19A1 as a hypoxia–immune-related gene in MM. Future functional and mechanistic studies will further clarify the roles of SLC19A1 in MM.
Collapse
Affiliation(s)
- Wenjin Li
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Peng Yuan
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Weiqin Liu
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Lichan Xiao
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Chun Xu
- Department of Hematology, Pingxiang People’s Hospital, Pingxiang, China
| | - Qiuyu Mo
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
- Department of Hematology, Second Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Shujuan Xu
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yuchan He
- Department of Hematology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Duanfeng Jiang
- Department of Hematology, Second Affiliated Hospital of Hainan Medical College, Haikou, China
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaotao Wang
- Department of Hematology, Affiliated Hospital of Guilin Medical University, Guilin, China
- *Correspondence: Xiaotao Wang,
| |
Collapse
|
29
|
Yan Q, Sun YS, An R, Liu F, Fang Q, Wang Z, Xu T, Chen L, Du J. Application and progress of the detection technologies in hepatocellular carcinoma. Genes Dis 2022. [PMID: 37492708 PMCID: PMC10363596 DOI: 10.1016/j.gendis.2022.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has a very high incidence and fatality rate, and in most cases, it is already at an advanced stage when diagnosed. Therefore, early prevention and detection of HCC are two of the most effective strategies. However, the methods recommended in the practice guidelines for the detection of HCC cannot guarantee high sensitivity and specificity except for the liver biopsy, which is known as the "gold standard". In this review, we divided the detection of HCC into pre-treatment diagnosis and post-treatment monitoring, and found that in addition to the traditional imaging detection and liver biopsy, alpha fetoprotein (AFP), lens culinaris-agglutinin-reactive fraction of AFP (AFP-L3), protein induced by vitamin K absence or antagonist-II (PIVKA-II) and other biomarkers are excellent biomarkers for HCC, especially when they are combined together. Most notably, the emerging liquid biopsy shows great promise in detecting HCC. In addition, lactic dehydrogenase (LDH), suppressor of cytokine signaling (SOCS) and other relevant biomarkers may become promising biomarkers for HCC post-treatment monitoring. Through the detailed introduction of the diagnostic technology of HCC, we can have a detailed understanding of its development process and then obtain some enlightenment from the diagnosis, to improve the diagnostic rate of HCC and reduce its mortality.
Collapse
|
30
|
Sfifou F, Ouzir M, Hakkou EM, Obtel M, Errihani H, Bouzidi AA, Abouqal R, El Ouahabi A, Cherradi N. Immunohistochemical expression of HIF-1α, IDH1 and TP53: Prognostic profile of Moroccan patients with diffuse glioma. J Chem Neuroanat 2021; 119:102056. [PMID: 34871733 DOI: 10.1016/j.jchemneu.2021.102056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/29/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
Abstract
Diffuse gliomas are growing brain tumors that occur in adult life. This study was designed to determine whether the immunohistochemical analysis of IDH1, HIF-1alpha, or TP53 can provide useful biomarkers of clinical severity and progression of diffuse gliomas. Also, it is hypothesized that the expression of IDH1 mutant induces HIF-1alpha. Immunohistochemical staining for HIF-1alpha, IDH1, and TP 53 was performed in biopsy or resection (sub-total or gross-total) tissue from diffuse gliomas in a clinical series of 32 patients. Associations of the HIF-1alpha, IDH1, and TP53 with clinical characteristics were evaluated and the co-expression of two biomarkers (HIF-1alpha and IDH1) was tested. Our data revealed that each biomarker is expressed in a subset of gliomas (IDH1 was positive in 56% cases, HIF-1alpha was positive in 50% cases and TP53 was positive in 44% cases). While no associations were found between clinical characteristics and the expression of HIF-1alpha, and TP53, IDH1 expression was associated with less severe clinical presentation (Karnofsky Performance Status) and disease progression and was more often expressed in females than males. In addition, there was no clear association between IDH1 and HIF-1alpha expression (21.9% of patients co-expressed IDH1 and HIF-1alpha). The current series provides clinical and immunohistochemical findings that can be useful for the clinical management of patients with diffused gliomas.
Collapse
Affiliation(s)
- Fatima Sfifou
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy, Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco.
| | - Mounir Ouzir
- Group of Research in Physiology and Physiopathology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, BP 1014 Rabat, Morocco
| | - El Mehdi Hakkou
- Neurosurgery Department, Hospital of Specialities in Rabat, Morocco
| | - Majdouline Obtel
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Hassan Errihani
- National Oncology Centre Sidi Mohamed Ben Abdallah in Rabat, Morocco
| | - Abderrahmane Al Bouzidi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy, Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Redouane Abouqal
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Rabat Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | | | - Nadia Cherradi
- Research's Pedagogic Unit of Pathological Anatomy, Laboratory of Pathological Anatomy, Research Team in Tumour Pathology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Pathological Anatomy Department, Hospital of Specialities in Rabat, Morocco
| |
Collapse
|
31
|
Jiang X, He Y, Shen Q, Duan L, Yuan Y, Tang L, Shi Y, Liu B, Zhai H, Shi P, Yang C, Chen Y. RETSAT Mutation Selected for Hypoxia Adaptation Inhibits Tumor Growth. Front Cell Dev Biol 2021; 9:744992. [PMID: 34805153 PMCID: PMC8601408 DOI: 10.3389/fcell.2021.744992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Hypoxia occurs not only in natural environments including high altitude, underground burrows and deep sea, but also in human pathological conditions, such as hypoxic solid tumors. It has been well documented that hypoxia related signaling pathway is associated with a poor clinical outcome. Our group has recently identified multiple novel genes critical for solid tumor growth comparing the genome-wide convergent/parallel sequence evolution of highland mammals. Among them, a single mutation on the retinol saturase gene (RETSAT) containing amino acid switch from glutamine (Q) to arginine (R) at the position 247 was identified. Here, we demonstrate that RETSAT is mostly downregulated in multiple types of human cancers, whose lower expression correlates with worse clinical outcome. We show that higher expression of RETSAT is positively associated with immune infiltration in different human cancers. Furthermore, we identify that the promoter region of RETSAT is highly methylated, which leads to its decreased expressions in tumor tissues comparing to normal tissues. Furthermore, we show that RETSAT knockdown promotes, while its overexpression inhibits, the cell proliferation ability of mouse embryonic fibroblasts (MEFs) and B16 in vitro. In addition, the mice carrying homozygous Q247R mutation (RETSATR/R) is more resistant to xenograft tumor formation, as well as DMBA/TPA induced cutaneous keratinocyte carcinoma formation, compared to littermate wild-type (RETSATQ/Q) mice. Mechanistic study uncovers that the oncogenic factor, the prolyl isomerase (PPIase) Pin1 and its related downstream signaling pathway, were both markedly repressed in the mutant mice compared to the wild-type mice. In summary, these results suggest that interdisciplinary study between evolution and tumor biology can facilitate identification of novel molecular events essential for hypoxic solid tumor growth in the future.
Collapse
Affiliation(s)
- Xiulin Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yaomei He
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qiushuo Shen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
| | - Lincan Duan
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yixiao Yuan
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lin Tang
- The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yulin Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Baiyang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Haoqing Zhai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Peng Shi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Cuiping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China.,Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
32
|
Jo MJ, Kim BG, Kim WY, Lee DH, Yun HK, Jeong S, Park SH, Kim BR, Kim JL, Kim DY, Lee SI, Oh SC. Cannabidiol Suppresses Angiogenesis and Stemness of Breast Cancer Cells by Downregulation of Hypoxia-Inducible Factors-1α. Cancers (Basel) 2021; 13:cancers13225667. [PMID: 34830821 PMCID: PMC8616476 DOI: 10.3390/cancers13225667] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Cannabidiol (CBD), one of the compounds present in the marijuana plant, has antitumor properties. However, the effect of CBD on breast cancer remains unclear. The aim of this study was to assess the effects of CBD for the angiogenesis and stemness of breast cancer cells by decreasing the expression of hypoxia-induced factor-1α (HIF-1α) through the Src/von Hippel–Lindau tumor suppressor protein (VHL) interaction. CBD can suppress angiogenesis and stem cell-like properties of breast cancer through Src/VHL/HIF-1α signaling. Abstract To assess the effect of Cannabidiol (CBD) on the angiogenesis and stemness of breast cancer cells as well as proliferation. Methods: mRNA level and the amount of protein of vascular endothelial growth factor (VEGF) were determined by qRT-PCR and ELISA. The angiogenic potential of breast cancer cells under hypoxic conditions was identified by the HUVEC tube formation assay. The degradation of HIF-1α by CBD and the Src/von Hippel–Lindau tumor suppressor protein (VHL) interaction were assessed by a co-immunoprecipitation assay and Western blotting. To identify the stemness of mamospheres, they were evaluated by the sphere-forming assay and flow cytometry. Results: CBD can suppress angiogenesis and stem cell-like properties of breast cancer through Src/VHL/HIF-1α signaling. CBD may potentially be utilized in the treatment of refractory or recurrent breast cancer.
Collapse
Affiliation(s)
- Min Jee Jo
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
| | - Bu Gyeom Kim
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
| | - Woo Young Kim
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea;
- Correspondence: (W.Y.K.); (D.-H.L.); Tel.: +82-2-2626-3078 (W.Y.K.); +82-33-640-2347 (D.-H.L.)
| | - Dae-Hee Lee
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangwon 25457, Korea
- Correspondence: (W.Y.K.); (D.-H.L.); Tel.: +82-2-2626-3078 (W.Y.K.); +82-33-640-2347 (D.-H.L.)
| | - Hye Kyeong Yun
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
| | - Soyeon Jeong
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea; (S.J.); (B.R.K.); (J.L.K.)
| | - Seong Hye Park
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
| | - Bo Ram Kim
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea; (S.J.); (B.R.K.); (J.L.K.)
| | - Jung Lim Kim
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea; (S.J.); (B.R.K.); (J.L.K.)
| | - Dae Yeong Kim
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
| | - Sun Il Lee
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea;
| | - Sang Cheul Oh
- Graduate School of Medicine, College of Medicine, Korea University, Seoul 08308, Korea; (M.J.J.); (B.G.K.); (H.K.Y.); (S.H.P.); (D.Y.K.); (S.C.O.)
- Department of Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea; (S.J.); (B.R.K.); (J.L.K.)
| |
Collapse
|
33
|
Deng X, Song Q, Zhang Y, Liu W, Hu H, Zhang Y. Tumour microenvironment-responsive nanoplatform based on biodegradable liposome-coated hollow MnO 2 for synergistically enhanced chemotherapy and photodynamic therapy. J Drug Target 2021; 30:334-347. [PMID: 34709119 DOI: 10.1080/1061186x.2021.1999961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Existing therapeutic efficacy of chemotherapy and photodynamic therapy (PDT) is always affected by some resistance factors from tumour environment (TME), such as hypoxia and the antioxidant defense system. PURPOSE This study aims at developing a cascaded intelligent multifunctional nanoplatforms to modulate the TME resistance for synergistically enhanced chemo- and photodynamic therapies. METHODS In this study, we synthesised hollow manganese dioxide nanoparticles (HMDNs) loaded with the hydrophilic chemotherapeutic drug (acriflavine, ACF) and the hydrophobic photosensitizer (chlorine6, Ce6), which was further encapsulated by pH-sensitive liposome to form core-shell nanocomposite, with surface modified with arginine-glycine-aspartic acid (RGD) peptide to achieve tumour targeting. RESULTS After uptake by tumour cells, the liposome shell was rapidly degraded by the low pH, and the inner core could be released from the liposome. Then, the released HMDNs/ACF/Ce6 would be dissociated by low pH and high levels of intracellular GSH within TME to release encapsulated drugs, thereby resulting in synergistic effects of chemotherapy and PDT. Meanwhile, the released ACF could bind with HIF-1a and then inhibit the expression levels of HIF-1's downstream signalling molecules P-gp and VEGF, which could further strengthen the antitumor effects. As a result, HMDNs/ACF/Ce6@Lipo-RGD NPs with laser irradiation exhibited superior anti-tumour therapeutic efficiency.
Collapse
Affiliation(s)
- Xiangtian Deng
- School of Medicine, Nankai University, Tianjin, China.,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Qingcheng Song
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Yiran Zhang
- School of Medicine, Nankai University, Tianjin, China.,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingze Zhang
- School of Medicine, Nankai University, Tianjin, China.,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| |
Collapse
|
34
|
Abstract
The proliferation, metastasis and therapy response of tumour cells are tightly regulated by interaction among various signalling networks. The microRNAs (miRNAs) can bind to 3'-UTR of mRNA and down-regulate expression of target gene. The miRNAs target various molecular pathways in regulating biological events such as apoptosis, differentiation, angiogenesis and migration. The aberrant expression of miRNAs occurs in cancers and they have both tumour-suppressor and tumour-promoting functions. On the contrary, SOX proteins are capable of binding to DNA and regulating gene expression. SOX2 is a well-known member of SOX family that its overexpression in different cancers to ensure progression and stemness. The present review focuses on modulatory impact of miRNAs on SOX2 in affecting growth, migration and therapy response of cancers. The lncRNAs and circRNAs can function as upstream mediators of miRNA/SOX2 axis in cancers. In addition, NF-κB, TNF-α and SOX17 are among other molecular pathways regulating miRNA/SOX2 axis in cancer. Noteworthy, anti-cancer compounds including bufalin and ovatodiolide are suggested to regulate miRNA/SOX2 axis in cancers. The translation of current findings to clinical course can pave the way to effective treatment of cancer patients and improve their prognosis.
Collapse
|
35
|
Burtscher J, Millet GP, Renner-Sattler K, Klimont J, Hackl M, Burtscher M. Moderate Altitude Residence Reduces Male Colorectal and Female Breast Cancer Mortality More Than Incidence: Therapeutic Implications? Cancers (Basel) 2021; 13:cancers13174420. [PMID: 34503229 PMCID: PMC8430507 DOI: 10.3390/cancers13174420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Living at moderate altitude may be associated with health benefits, including reduced mortality from male colorectal and female breast cancer. We aimed to determine altitude-dependent incidence and mortality rates of those cancers and put them in the context of altitude-associated lifestyle differences. METHODS Incidence cases and deaths of male colorectal cancer (n = 17,712 and 7462) and female breast cancer (n = 33,803 and 9147) from altitude categories between 250 to about 2000 m were extracted from official Austrian registries across 10 years (2008-2017). Altitude-associated differences in health determinants were derived from the Austrian Health Interview Survey (2014). RESULTS The age-standardized incidence and mortality rates of male colorectal cancer decreased by 24.0% and 44.2%, and that of female breast cancer by 6.5% and 26.2%, respectively, from the lowest to the highest altitude level. Higher physical activity levels and lower body mass index for both sexes living at higher altitudes were found. CONCLUSIONS Living at a moderate altitude was associated with a reduced incidence and (more pronounced) mortality from colorectal and breast cancer. Our results suggest a complex interaction between specific climate conditions and lifestyle behaviours. These observations may, in certain cases, support decision making when changing residence.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Biomedical Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (J.B.); (G.P.M.)
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland
| | - Grégoire P. Millet
- Department of Biomedical Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland; (J.B.); (G.P.M.)
- Institute of Sport Sciences, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | - Jeannette Klimont
- Unit Demography and Health, Directorate Social Statistics, Statistics Austria, 1110 Vienna, Austria;
| | - Monika Hackl
- Austrian National Cancer Registry, Directorate Social Statistics, Statistics Austria, 1110 Vienna, Austria;
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, A-6020 Innsbruck, Austria
- Correspondence:
| |
Collapse
|
36
|
Shenouda S, Kulkarni K, Abuetabh Y, Sergi C. Cancer Stem Cells and their Management in Cancer Therapy. Recent Pat Anticancer Drug Discov 2021; 15:212-227. [PMID: 32660407 DOI: 10.2174/1574892815666200713145931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/16/2020] [Accepted: 06/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In the last decade, the proposed Cancer Stem Cell (CSC) hypothesis has steadily changed the way cancer treatment is approached. CSCs may be the source of the heterogeneous non-tumorigenic cell population included in a neoplasm. Intratumor and intertumoral heterogeneity is a well-known phenomenon that massively entangles the diagnosis and treatment of cancer. The literature seems to suggest that heterogeneity develops progressively within tumor-initiating stem cells. CSCs harbor genetic and/or epigenetic alterations that allow them to differentiate into multiple tumor cell types sequentially. OBJECTIVE The CSC hypothesis, cellular therapy, and the most recent patents on CSCs were reviewed. METHODS PubMed, Scopus, and Google Scholar were screened for this information. Also, an analysis of the most recent data targeting CSCs in pediatric cancer developed at two Canadian institutions is provided. The genes involved with the activation of CSCs and the drugs used to antagonize them are also highlighted. RESULTS It is underlined that (1) CSCs possess stem cell-like properties, including the ability for self-renewal; (2) CSCs can start carcinogenesis and are responsible for tumor recurrence after treatment; (3) Although some limitations have been raised, which may oppose the CSC hypothesis, cancer progression and metastasis have been recognized to be caused by CSCs. CONCLUSION The significant roles of cell therapy may include an auto-transplant with high-dose treatment, an improvement of the immune function, creation of chimeric antigen receptor T cells, and the recruitment of NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Suzan Shenouda
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Ketan Kulkarni
- Department of Pediatrics, Pediatric Hematology/Oncology, Halifax, NS, Canada
| | - Yasser Abuetabh
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Consolato Sergi
- Department of Lab. Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
37
|
A Hypoxia Gene-Based Signature to Predict the Survival and Affect the Tumor Immune Microenvironment of Osteosarcoma in Children. J Immunol Res 2021; 2021:5523832. [PMID: 34337075 PMCID: PMC8299210 DOI: 10.1155/2021/5523832] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/09/2021] [Accepted: 07/05/2021] [Indexed: 01/15/2023] Open
Abstract
Osteosarcoma is a quickly developing, malignant cancer of the bone, which is associated with a bad prognosis. In osteosarcoma, hypoxia promotes the malignant phenotype, which results in a cascade of immunosuppressive processes, poor prognosis, and a high risk of metastasis. Nonetheless, additional methodologies for the study of hyperoxia in the tumor microenvironment also need more analysis. We obtained 88 children patients with osteosarcoma from the Therapeutically Applicable Research to Generate Effective Treatment (TARGET) database and 53 children patients with RNA sequence and clinicopathological data from the Gene Expression Omnibus (GEO). We developed a four-gene signature related to hypoxia to reflect the immune microenvironment in osteosarcoma that predicts survival. A high-risk score indicated a poor prognosis and immunosuppressive microenvironment. The presence of the four-gene signature related to hypoxia was correlated with clinical and molecular features and was an important prognostic predictor for pediatric osteosarcoma patients. In summary, we established and validated a four-gene signature related to hypoxia to forecast recovery and presented an independent prognostic predictor representing overall immune response strength within the osteosarcoma microenvironment.
Collapse
|
38
|
Hypoxia in Lung Cancer Management: A Translational Approach. Cancers (Basel) 2021; 13:cancers13143421. [PMID: 34298636 PMCID: PMC8307602 DOI: 10.3390/cancers13143421] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hypoxia is a common feature of lung cancers. Nonetheless, no guidelines have been established to integrate hypoxia-associated biomarkers in patient management. Here, we discuss the current knowledge and provide translational novel considerations regarding its clinical detection and targeting to improve the outcome of patients with non-small-cell lung carcinoma of all stages. Abstract Lung cancer represents the first cause of death by cancer worldwide and remains a challenging public health issue. Hypoxia, as a relevant biomarker, has raised high expectations for clinical practice. Here, we review clinical and pathological features related to hypoxic lung tumours. Secondly, we expound on the main current techniques to evaluate hypoxic status in NSCLC focusing on positive emission tomography. We present existing alternative experimental approaches such as the examination of circulating markers and highlight the interest in non-invasive markers. Finally, we evaluate the relevance of investigating hypoxia in lung cancer management as a companion biomarker at various lung cancer stages. Hypoxia could support the identification of patients with higher risks of NSCLC. Moreover, the presence of hypoxia in treated tumours could help clinicians predict a worse prognosis for patients with resected NSCLC and may help identify patients who would benefit potentially from adjuvant therapies. Globally, the large quantity of translational data incites experimental and clinical studies to implement the characterisation of hypoxia in clinical NSCLC management.
Collapse
|
39
|
Upadhyay N, Tilekar K, Safuan S, Kumar AP, Stalin J, Ruegg C, Ramaa C S. Recent Anti‐angiogenic Drug Discovery Efforts To Combat Cancer. ChemistrySelect 2021. [DOI: 10.1002/slct.202101792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Neha Upadhyay
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| | - Kalpana Tilekar
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| | - Sabreena Safuan
- Pusat pengajian sains School of Health Sciences Universiti Sains Malaysia Malaysia 16150 Kubang Kerian Kelantan
| | - Alan P. Kumar
- Department of Pharmacology National University of Singapore Singapore
| | - Jimmy Stalin
- Department of Oncology Microbiology, and Immunology University of Fribourg Chemin du Musée 18, PER17, CH 1700 Fribourg Switzerland
| | - Curzio Ruegg
- Department of Oncology Microbiology, and Immunology University of Fribourg Chemin du Musée 18, PER17, CH 1700 Fribourg Switzerland
| | - Ramaa C S
- Department of Pharmaceutical Chemistry Bharati Vidyapeeth's College of Pharmacy Sector 8, CBD Belapur Navi Mumbai 400614 India
| |
Collapse
|
40
|
Mohan A, Raj Rajan R, Mohan G, Kollenchery Puthenveettil P, Maliekal TT. Markers and Reporters to Reveal the Hierarchy in Heterogeneous Cancer Stem Cells. Front Cell Dev Biol 2021; 9:668851. [PMID: 34150761 PMCID: PMC8209516 DOI: 10.3389/fcell.2021.668851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
A subpopulation within cancer, known as cancer stem cells (CSCs), regulates tumor initiation, chemoresistance, and metastasis. At a closer look, CSCs show functional heterogeneity and hierarchical organization. The present review is an attempt to assign marker profiles to define the functional heterogeneity and hierarchical organization of CSCs, based on a series of single-cell analyses. The evidences show that analogous to stem cell hierarchy, self-renewing Quiescent CSCs give rise to the Progenitor CSCs with limited proliferative capacity, and later to a Progenitor-like CSCs, which differentiates to Proliferating non-CSCs. Functionally, the CSCs can be tumor-initiating cells (TICs), drug-resistant CSCs, or metastasis initiating cells (MICs). Although there are certain marker profiles used to identify CSCs of different cancers, molecules like CD44, CD133, ALDH1A1, ABCG2, and pluripotency markers [Octamer binding transcriptional factor 4 (OCT4), SOX2, and NANOG] are used to mark CSCs of a wide range of cancers, ranging from hematological malignancies to solid tumors. Our analysis of the recent reports showed that a combination of these markers can demarcate the heterogeneous CSCs in solid tumors. Reporter constructs are widely used for easy identification and quantification of marker molecules. In this review, we discuss the suitability of reporters for the widely used CSC markers that can define the heterogeneous CSCs. Since the CSC-specific functions of CD44 and CD133 are regulated at the post-translational level, we do not recommend the reporters for these molecules for the detection of CSCs. A promoter-based reporter for ABCG2 may also be not relevant in CSCs, as the expression of the molecule in cancer is mainly regulated by promoter demethylation. In this context, a dual reporter consisting of one of the pluripotency markers and ALDH1A1 will be useful in marking the heterogeneous CSCs. This system can be easily adapted to high-throughput platforms to screen drugs for eliminating CSCs.
Collapse
Affiliation(s)
- Amrutha Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Manipal Academy of Higher Education, Manipal, India
| | - Reshma Raj Rajan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | | |
Collapse
|
41
|
Luo X, Li A, Chi X, Lin Y, Liu X, Zhang L, Su X, Yin Z, Lin H, Gao J. Hypoxia-Activated Prodrug Enabling Synchronous Chemotherapy and HIF-1α Downregulation for Tumor Treatment. Bioconjug Chem 2021; 32:983-990. [PMID: 33847488 DOI: 10.1021/acs.bioconjchem.1c00131] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The overexpression of HIF-1α in solid tumors due to hypoxia is closely related to drug resistance and consequent treatment failure. Herein, we constructed a hypoxia-activated prodrug named as YC-Dox. This prodrug could be activated under hypoxic conditions and undergo self-immolation to release doxorubicin (Dox) and YC-1 hemisuccinate (YCH-1), which could execute chemotherapy and result in HIF-1α downregulation, respectively. This prodrug is capable of specifically releasing Dox and YCH-1 in response to hypoxia, leading to a substantial synergistic potency and a remarkable cytotoxic selectivity (>8-fold) for hypoxic cancer cells over normoxic healthy cells. The in vivo experiments reveal that this prodrug can selectively aim at hypoxic cancer cells and avoid undesired targeting of normal cells, leading to elevated therapeutic efficacy for tumor treatment and minimized adverse effects on normal tissues.
Collapse
Affiliation(s)
- Xiangjie Luo
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Ao Li
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xiaoqin Chi
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Yaying Lin
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Xing Liu
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Lifan Zhang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Xinhui Su
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Zhenyu Yin
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen 361004, China
| | - Hongyu Lin
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jinhao Gao
- The Key Laboratory for Chemical Biology of Fujian Province, The MOE Laboratory of Spectrochemical Analysis & Instrumentation, and Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
42
|
Mohan A, Raj R R, Mohan G, K P P, Thomas Maliekal T. Reporters of Cancer Stem Cells as a Tool for Drug Discovery. Front Oncol 2021; 11:669250. [PMID: 33968778 PMCID: PMC8100607 DOI: 10.3389/fonc.2021.669250] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/29/2021] [Indexed: 01/04/2023] Open
Abstract
In view of the importance of cancer stem cells (CSCs) in chemoresistance, metastasis and recurrence, the biology of CSCs were explored in detail. Based on that, several modalities were proposed to target them. In spite of the several clinical trials, a successful CSC-targeting drug is yet to be identified. The number of molecules screened and entered for clinical trial for CSC-targeting is comparatively low, compared to other drugs. The bottle neck is the lack of a high-throughput adaptable screening strategy for CSCs. This review is aimed to identify suitable reporters for CSCs that can be used to identify the heterogeneous CSC populations, including quiescent CSCs, proliferative CSCs, drug resistant CSCs and metastatic CSCs. Analysis of the tumor microenvironment regulating CSCs revealed that the factors in CSC-niche activates effector molecules that function as CSC markers, including pluripotency markers, CD133, ABCG2 and ALDH1A1. Among these factors OCT4, SOX2, NANOG, ABCG2 and ALDH1A1 are ideal for making reporters for CSCs. The pluripotency molecules, like OCT4, SOX2 and NANOG, regulate self-renewal, chemoresistance and metastasis. ABCG2 is a known regulator of drug resistance while ALDH1A1 modulates self-renewal, chemoresistance and metastasis. Considering the heterogeneity of CSCs, including a quiescent population and a proliferative population with metastatic ability, we propose the use of a combination of reporters. A dual reporter consisting of a pluripotency marker and a marker like ALDH1A1 will be useful in screening drugs that target CSCs.
Collapse
Affiliation(s)
- Amrutha Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India.,Centre for Doctoral Studies, Manipal Academy of Higher Education, Manipal, India
| | - Reshma Raj R
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Gayathri Mohan
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Padmaja K P
- Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | |
Collapse
|
43
|
Calderón-Gerstein WS, Torres-Samaniego G. High altitude and cancer: An old controversy. Respir Physiol Neurobiol 2021; 289:103655. [PMID: 33766749 DOI: 10.1016/j.resp.2021.103655] [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: 08/05/2020] [Revised: 03/05/2021] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
Ecological studies have found that individuals that live at high altitude regions and in places where ultraviolet radiation is maximal, have lower rates of different types of cancer. However, there is evidence that in these same regions, genetic mutations that are prooncogenic, develop, as they are needed to increase human adaptability to hypoxic environments. Debate has arisen between researchers who consider high altitude environments as suitable for human longevity because of its protective effects against malignancies, and scientists that have reported an increased incidence of different type of cancers in these same regions. Evidence is presented that altitude is related to the development of genetic alterations in micro RNAs, p53 protein, lymphocyte activity, decrease in Fas Ligand and other proapoptotic molecules, as well as increase in prometastatic VGEF an HIF. Notwithstanding, higher vitamin D and ultraviolet B levels, and a better metabolic profile, taken together with lower pollution levels hves been related to lower incidence and mortality rates from malignancies in a series of epidemiological studies.
Collapse
Affiliation(s)
- Walter S Calderón-Gerstein
- Universidad Continental, Huancayo, Peru; EsSalud Junín, Peru; Altitude Medicine Research Association (ASIMEDIAL), Peru.
| | - Gabriela Torres-Samaniego
- San Pedro de Cajas Health Post, EsSalud, Tarma, Peru; Altitude Medicine Research Association (ASIMEDIAL), Peru
| |
Collapse
|
44
|
Zeng Z, Lei S, He Z, Chen T, Jiang J. YEATS2 is a target of HIF1α and promotes pancreatic cancer cell proliferation and migration. J Cell Physiol 2021; 236:2087-2098. [PMID: 32749678 DOI: 10.1002/jcp.29995] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 07/17/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022]
Abstract
Hypoxia is involved in the development of pancreatic cancer (PC). The responses of hypoxia-associated genes and their regulated mechanisms are largely unknown. In this study, through bioinformatic analysis and quantitative real-time polymerase chain reaction, the YEATS domain containing 2 (YEATS2) was determined to be a key hypoxia-associated gene. It was increased in PC cells under hypoxia, upregulated in PC tissues, and predicted poor outcome. YEATS2 inhibition decreased the proliferation and migration of PC cells under both normoxia and hypoxia in vitro as well as proliferation and metastasis in vivo. We found that hypoxia-inducible factor 1α (HIF1α) regulated the expression of YEATS2 via binding to the hypoxia response element (HRE) of YEATS2 and coexpressed with YEATS2 in PC tissues. Overexpression of YEATS2 blocked the inhibitory effects of HIF1α silence on PC cell proliferation and migration under hypoxia. Collectively, our study revealed that YEATS2 is a target gene of HIF1α and promotes PC development under hypoxia.
Collapse
Affiliation(s)
- Zhirui Zeng
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Tissue Engineering and Stem Cell of Guizhou Province, Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Shan Lei
- Key Laboratory of Tissue Engineering and Stem Cell of Guizhou Province, Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhiwei He
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tengxiang Chen
- Key Laboratory of Tissue Engineering and Stem Cell of Guizhou Province, Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jianxin Jiang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease of Wuhan University, Wuhan, China
| |
Collapse
|
45
|
Li Y, Sun P, Zhao L, Yan X, Ng DKP, Lo P. Ferric Ion Driven Assembly of Catalase‐like Supramolecular Photosensitizing Nanozymes for Combating Hypoxic Tumors. Angew Chem Int Ed Engl 2020; 59:23228-23238. [DOI: 10.1002/anie.202010005] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/24/2020] [Indexed: 01/28/2023]
Affiliation(s)
- Yongxin Li
- Department of Biomedical Sciences City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| | - Pan Sun
- CAS Key Laboratory of Green Process and Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Luyang Zhao
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Dennis K. P. Ng
- Department of Chemistry The Chinese University of Hong Kong Shatin N.T. Hong Kong China
| | - Pui‐Chi Lo
- Department of Biomedical Sciences City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| |
Collapse
|
46
|
Li Y, Sun P, Zhao L, Yan X, Ng DKP, Lo P. Ferric Ion Driven Assembly of Catalase‐like Supramolecular Photosensitizing Nanozymes for Combating Hypoxic Tumors. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202010005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yongxin Li
- Department of Biomedical Sciences City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| | - Pan Sun
- CAS Key Laboratory of Green Process and Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Luyang Zhao
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences Beijing 100190 China
| | - Dennis K. P. Ng
- Department of Chemistry The Chinese University of Hong Kong Shatin N.T. Hong Kong China
| | - Pui‐Chi Lo
- Department of Biomedical Sciences City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong China
| |
Collapse
|
47
|
Terry S, Engelsen AST, Buart S, Elsayed WS, Venkatesh GH, Chouaib S. Hypoxia-driven intratumor heterogeneity and immune evasion. Cancer Lett 2020; 492:1-10. [PMID: 32712233 DOI: 10.1016/j.canlet.2020.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/24/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022]
Abstract
While it is widely accepted that high intratumoral heterogeneity confers serious challenges in the emerging resistance and the subsequent effective therapeutic targeting of cancer, the underlying biology of intratumoral heterogeneity remains elusive. In particular, it remains to be fully elucidated how microenvironmental factors shape genetic and non-genetic heterogeneity, which in turn determine the course of tumor evolution and clinical progression. In this context, hypoxia, a hallmark of most growing cancers, characterized by decreased O2 partial pressure is a key player of the tumor microenvironment. Despite extensive data indicating that hypoxia promotes cellular metabolic adaptation, immune suppression and various steps of tumor progression via hypoxia regulated gene transcription, much less is known about the role of hypoxia in mediating therapy resistance as a driver of tumor evolution through genetic and non-genetic mechanisms. In this review, we will discuss recent evidence supporting a prominent role of hypoxia as a driver of tumor heterogeneity and highlight the multifaceted manner by which this in turn could impact cancer evolution, reprogramming and immune escape. Finally, we will discuss how detailed knowledge of the hypoxic footprint may open up new therapeutic avenues for the management of cancer.
Collapse
Affiliation(s)
- Stéphane Terry
- INSERM UMR 1186, Integrative Tumour Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France; Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Agnete S T Engelsen
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway.
| | - Stéphanie Buart
- INSERM UMR 1186, Integrative Tumour Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France.
| | - Walid Shaaban Elsayed
- Department of Oral Biology, College of Dentistry, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumour Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France; Thumbay Research Institute of Precision Medicine, Gulf Medical University, Ajman, 4184, United Arab Emirates.
| |
Collapse
|