1
|
Tian Q, Zhu Z, Feng Y, Zhao S, Lin H, Zhang W, Xu Z. H 2O 2-activated mitochondria-targeting photosensitizer for fluorescence imaging-guided combination photodynamic and radiotherapy. J Mater Chem B 2024; 13:326-335. [PMID: 39552242 DOI: 10.1039/d4tb01653j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Radiotherapy is a primary modality in cancer treatment but is accompanied by severe side effects to healthy tissues and radiation resistance to some extent. To overcome these limitations, we developed a H2O2-responsive photosensitizer, CyBT, which could be activated by the upregulated H2O2 induced by radiotherapy, enabling near-infrared fluorescence imaging-guided combination photodynamic and radiotherapy. The synthesis of CyBT began with the covalent linkage of hemicyanine and a free radical TEMPO through the click reaction, which demonstrated superior photodynamic properties. Shielding of fluorescence and photodynamic activity was achieved by incorporating phenylboronic acid pinacol ester. In X-ray irradiated tumor cells, the upregulation of H2O2 activated CyBT, thereby restoring its fluorescence and photodynamic activity. Additionally, the positive charge of CyBT facilitated its targeting to the mitochondria within tumor cells for more efficiently triggering cell apoptosis. CyBT was co-assembled with a polymer PEG-b-PDPA to form acid-responsive nanoparticles (NPs-CyBT). This formulation enhanced tumor targeting, improved water solubility of CyBT, and extended in vivo circulation time. Utilizing fluorescence imaging to guide photodynamic and radiotherapy, NPs-CyBT can accurately target solid tumors in mice, and lead to tumor elimination, suggesting that it is a potential strategy for the effective treatment of malignant tumors.
Collapse
Affiliation(s)
- Qiufen Tian
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Zifan Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Yun Feng
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Shirui Zhao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Hui Lin
- Gastroenterology department, Shanghai Jing'an District Zhabei Central Hospital, Shanghai 200072, China.
| | - Wen Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
2
|
Cheng YT, Chang DM, Tung YC, Hsiao PW, Nakagawa-Goto K, Shyur LF. Phytosesquiterpene lactones deregulate mitochondrial activity and phenotypes associated with triple-negative breast cancer metastasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156226. [PMID: 39571415 DOI: 10.1016/j.phymed.2024.156226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) recurrence and metastasis are the major causes of failure in TNBC therapy. The difficulties in treating TNBCs may be because of increased cancer cell plasticity that involves the fine-tuning of cellular redox homeostasis, mitochondrial bioenergetics, metabolic characteristics, and the development of cancer stem cells (CSCs). PURPOSE To investigate the effects and the underlying mechanisms of the phytosesquiterpene lactone deoxyelephantopin (DET) and its semi-synthesized derivative (DETD-35) in suppressing different phenotypic TNBC cell populations that contribute to tumor metastasis. METHODS A timelapse microfluidic-based system was established to analyze the effects of DETD-35 and DET on cell migration behavior in an oxygen gradient. Seahorse real-time cell metabolic analyzer and gas chromatography/quadrupole-time-of-flight mass spectrometry (GC/Q-TOF MS) were utilized to analyze the effects of the compounds on mitochondrial bioenergetics in TNBC cells. A miRNA knockout technique and miRNA sponges were employed to evaluate the miR-4284 involvement in the anti-TNBC cell effect of either compound. RESULTS DETD-35 and DET attenuated TNBC cell migration toward hypoxic regions under a 2-19 % oxygen gradient in a timelapse microfluidic-based system. DETD-35 and DET also suppressed CSC-like phenotypes, including the expression of Sox2, Oct4, and CD44 in TNBC cells under hypoxic conditions. DETD-35 and DET affected mitochondrial basal respiration, ATP production, proton leak, and primary metabolism, including glycolysis, the TCA cycle, and amino acid metabolism in the lung-metastatic TNBC cells. Furthermore, the expression of mitophagy markers PARKIN, BNIP3, PINK1, LC3-II, and apoptotic markers Bax, cleaved caspase 7, and cleaved PARP in hypoxic and lung-metastatic TNBC cells was also regulated by treatment with either compound. In miR-4284 knockout cells or miR-4284 inhibitor co-treated TNBC cells, DET- and DETD-35-induced over-expression of mitophagic and apoptotic markers was partially reversed, indicating miR-4284 involved with the compounds caused programmed cell death. CONCLUSION This study demonstrated the novel activities of DETD-35 and DET in suppressing CSC-like phenotypes and metastatic TNBC cells through the de-regulation of mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Yu-Ting Cheng
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan; Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Dao-Ming Chang
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei, 11529, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Kyoko Nakagawa-Goto
- College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Lie-Fen Shyur
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica, Taipei 11529, Taiwan; Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan; Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan; Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei 11031, Taiwan; PhD Program in Translational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
3
|
Kato T, Sugihara E, Hata Y, Kawakami K, Fujita Y, Mizutani K, Ando T, Sakai Y, Sakurai K, Toyota S, Ehara H, Ito M, Ito H. Diagnostic potential of SDHB mRNA contained in serum extracellular vesicles among patients with prostate cancer. Prostate 2024; 84:1515-1524. [PMID: 39279231 DOI: 10.1002/pros.24792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Androgen receptor signaling inhibitors(ARSIs) have been used to treat patients with metastatic prostate cancer (PC) and castration-resistant prostate cancer (CRPC). In this study, we aimed to identify novel serum extracellular vesicle (EV)-based biomarkers to diagnose ARSI-resistance and therapeutic targets for ARSI-resistant CRPC. METHODS Total RNA contained in serum EVs from 5 cases of CRPC before ARSI treatment and after acquiring ARSI-resistance was subjected to RNA-sequencing. The expression changes of selected RNAs contained in EVs were confirmed in 48 cases of benign prostatic hyperplasia (BPH) and 107 PC using reverse transcription-quantitative PCR (RT-qPCR) and compared with tissue RNA expression using public datasets. RESULTS RNA-sequencing revealed that mitochondrial oxidative phosphorylation (OXPHOS)-related genes were increased in EVs after acquiring ARSI-resistance. Among them, RT-qPCR and datasets analysis demonstrated that SDHB mRNA was upregulated after acquiring ARSI-resistance in EVs and ARSI-exposed PC tissue compared to ARSI-naïve EVs and tissue, respectively. SDHB mRNA levels both in EVs and tissue were increased in localized PC compared with BPH and decreased in advanced PC. Tissue expression of SDHB mRNA was significantly correlated with those of other OXPHOS-related genes. SDHB mRNA in EVs (EV-SDHB) was elevated among 3 out of 7 ARSI-treating patients with stable PSA levels who later progressed to ARSI-resistant CRPC. CONCLUSIONS The levels of OXPHOS-related mRNAs in EVs correlated with those in PC tissue, among which SDHB mRNA was found to be a novel biomarker to diagnose ARSI-resistance. EV-SDHB may be useful for early diagnosis of ARSI-resistance.
Collapse
MESH Headings
- Humans
- Male
- Extracellular Vesicles/metabolism
- RNA, Messenger/blood
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/diagnosis
- Prostatic Neoplasms, Castration-Resistant/blood
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Succinate Dehydrogenase/genetics
- Succinate Dehydrogenase/metabolism
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Aged
- Middle Aged
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/diagnosis
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/blood
- Prostatic Neoplasms/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Drug Resistance, Neoplasm
- Prostatic Hyperplasia/genetics
- Prostatic Hyperplasia/blood
- Prostatic Hyperplasia/diagnosis
- Prostatic Hyperplasia/metabolism
- Androgen Antagonists/therapeutic use
- Androgen Antagonists/pharmacology
Collapse
Affiliation(s)
- Taku Kato
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Urology, Asahi University Hospital, Gifu, Japan
| | - Eiji Sugihara
- Open Facility Center, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yuko Hata
- Open Facility Center, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kyojiro Kawakami
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yasunori Fujita
- Molecular and Cellular Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kosuke Mizutani
- Department of Urology, Central Japan International Medical Center, Gifu, Japan
| | - Tatsuya Ando
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yasuhiro Sakai
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Kouhei Sakurai
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shohei Toyota
- Department of Urology, Asahi University Hospital, Gifu, Japan
| | - Hidetoshi Ehara
- Department of Urology, Asahi University Hospital, Gifu, Japan
| | - Masafumi Ito
- Research Team for Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hiroyasu Ito
- Department of Joint Research Laboratory of Clinical Medicine, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
4
|
Tang HP, Zhu EL, Bai QX, Wang S, Wang ZB, Wang M, Kuang HX. Polygala japonica Houtt.: A comprehensive review on its botany, traditional uses, phytochemistry, pharmacology, and pharmacokinetics. Fitoterapia 2024; 179:106233. [PMID: 39326795 DOI: 10.1016/j.fitote.2024.106233] [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: 07/07/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Polygala japonica Houtt. (P. japonica), a member of the Polygala genus in the Polygalaceae family, has been historically utilized in traditional folk medicine as an expectorant, anti-inflammatory, anti-bacterial, and anti-depressant agent. This paper systematically reviews the latest research in botany, traditional uses, phytochemistry, pharmacology, and pharmacokinetics, aiming to provide a scientific foundation for the future development and application of P. japonica and to explore its potential value comprehensively. Approximately 86 compounds have been isolated from P. japonica, with triterpenoid saponins being the most prevalent and bioactive components. Extensive pharmacological activities of P. japonica extracts or compounds have been confirmed in vivo and in vitro, including anti-inflammatory, anti-depressant, neuroprotective, anti-obesity, anti-apoptotic, and skin-protective effects. Additionally, P. japonica has demonstrated significant curative effects and relatively clear pharmacological mechanisms in treating inflammatory and nervous system diseases. Specific components of its primary triterpenoid saponins are rapidly absorbed in the body. This review advocates for deeper scientific research on P. japonica, noting that most current research remains in its early stages and many reported biological activities require further clinical validation. Despite this, the traditional medical use of P. japonica across various cultures attests to its broad application value. Presently, the pharmacological activities of P. japonica extracts and compounds provide a scientific basis for its traditional uses. Future research must ensure the safety and effectiveness of P. japonica through in-depth pharmacokinetic studies, and the establishment of a refined and standardized quality evaluation system is essential for its clinical development and application.
Collapse
Affiliation(s)
- Hai-Peng Tang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - En-Lin Zhu
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qian-Xiang Bai
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Shuang Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Zhi-Bin Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China
| | - Meng Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China.
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Harbin 150000, China.
| |
Collapse
|
5
|
Kamble OS, Chatterjee R, Abishek KG, Chandra J, Alsayari A, Wahab S, Sahebkar A, Kesharwani P, Dandela R. Small molecules targeting mitochondria as an innovative approach to cancer therapy. Cell Signal 2024; 124:111396. [PMID: 39251050 DOI: 10.1016/j.cellsig.2024.111396] [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: 07/16/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Cellular death evasion is a defining characteristic of human malignancies and a significant contributor to therapeutic inefficacy. As a result of oncogenic inhibition of cell death mechanisms, established therapeutic regimens seems to be ineffective. Mitochondria serve as the cellular powerhouses, but they also function as repositories of self-destructive weaponry. Changes in the structure and activities of mitochondria have been consistently documented in cancer cells. In recent years, there has been an increasing focus on using mitochondria as a targeted approach for treating cancer. Considerable attention has been devoted to the development of delivery systems that selectively aim to deliver small molecules called "mitocans" to mitochondria, with the ultimate goal of modulating the physiology of cancer cells. This review summarizes the rationale and mechanism of mitochondrial targeting with small molecules in the treatment of cancer, and their impact on the mitochondria. This paper provides a concise overview of the reasoning and mechanism behind directing treatment towards mitochondria in cancer therapy, with a particular focus on targeting using small molecules. This review also examines diverse small molecule types within each category as potential therapeutic agents for cancer.
Collapse
Affiliation(s)
- Omkar S Kamble
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Rana Chatterjee
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - K G Abishek
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India
| | - Jyoti Chandra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Indian Oil Odisha Campus, Samantpuri, Bhubaneswar 751013, India.
| |
Collapse
|
6
|
Zhou Q, You Y, Zhao Y, Xiao S, Song Z, Huang C, Qian J, Lu W, Tong H, Zhang Y, Wang Z, Li W, Zhang C, Guo X, Luo R, Hou Y, Cui J, Lu L, Zhou Y. TRPV4 drives the progression of leiomyosarcoma by promoting ECM1 generation and co-activating the FAK/PI3K/AKT/GSK3β pathway. Cell Oncol (Dordr) 2024:10.1007/s13402-024-01008-7. [PMID: 39612152 DOI: 10.1007/s13402-024-01008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2024] [Indexed: 11/30/2024] Open
Abstract
PURPOSE Leiomyosarcoma (LMS) is an aggressive mesenchymal malignant tumor with poor therapeutic options, but the molecular mechanisms underlying LMS remain largely unknown. Increasing evidence indicates that transient receptor potential vanilloid 4 (TRPV4) levels are closely related to the advancement of various malignant tumors through diverse molecular mechanisms. However, the roles and regulatory mechanisms of TRPV4 in LMS progression remain unclear. METHODS Immunohistochemistry, Western blot, and immunofluorescence were used to investigate the relationship between TRPV4 expression and LMS. Survival analysis was conducted to evaluate the association between TRPV4 levels and prognosis in LMS patients. Intracellular Ca2+ measurement, colony formation, CCK-8, wound healing and Transwell assays and peritoneal metastasis mouse model were used to verify the effect of TRPV4 activity and expression on LMS proliferation and metastasis. RNA-seq and proteomics were performed to explore the underlying mechanism. RESULTS TRPV4 was upregulated in LMS tissues and cells and served as a novel prognostic factor. Moreover, TRPV4 overexpression enhanced cell proliferation, cell migration and invasion of LMS cells in vitro, as well as promoted tumor metastasis in vivo, which could be blocked by HC067047 intervention or TRPV4 knockdown. Combined RNA-seq and proteomics analysis of KEGG pathway indicated that ECM receptor interaction was obviously activated. Extracellular matrix protein 1 (ECM1) was identified as downstream gene of TRPV4. Mechanistically, TRPV4 overexpression increased ECM1 level and activated the FAK/PI3K/AKT/GSK3β pathway, which could be reversed by TRPV4 knockdown or LY294002 treatment. Moreover, ECM1 overexpression enhanced the activation of FAK/PI3K/AKT/GSK3β pathway. And simultaneous overexpression of TRPV4 and ECM1 synergistically activated this pathway. CONCLUSION Our findings provide a novel mechanism by which TRPV4 directly activates Ca2+/FAK/PI3K/AKT/GSK3β pathway and further indirectly enhances the FAK/PI3K/AKT/GSK3β pathway through the promotion and secretion of ECM1 to promote LMS malignant progression. Targeting the TRPV4/FAK axis might be a promising potential strategy for prognosis and treatment of LMS.
Collapse
Affiliation(s)
- Qiwen Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yang You
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingying Zhao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
- Department of Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, 215002, China
| | - Shuxiu Xiao
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhengqing Song
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chuxin Huang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Weiqi Lu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hanxing Tong
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiming Wang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Li
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chenlu Zhang
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xi Guo
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, 200032, China
| | - Lili Lu
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Xiamen Key Laboratory of Biotherapy, Xiamen, 361000, China.
| | - Yuhong Zhou
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Centre for Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Al Khzem AH, Gomaa MS, Alturki MS, Tawfeeq N, Sarafroz M, Alonaizi SM, Al Faran A, Alrumaihi LA, Alansari FA, Alghamdi AA. Drug Repurposing for Cancer Treatment: A Comprehensive Review. Int J Mol Sci 2024; 25:12441. [PMID: 39596504 PMCID: PMC11595001 DOI: 10.3390/ijms252212441] [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: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer ranks among the primary contributors to global mortality. In 2022, the global incidence of new cancer cases reached about 20 million, while the number of cancer-related fatalities reached 9.7 million. In Saudi Arabia, there were 13,399 deaths caused by cancer and 28,113 newly diagnosed cases of cancer. Drug repurposing is a drug discovery strategy that has gained special attention and implementation to enhance the process of drug development due to its time- and money-saving effect. It involves repositioning existing medications to new clinical applications. Cancer treatment is a therapeutic area where drug repurposing has shown the most prominent impact. This review presents a compilation of medications that have been repurposed for the treatment of various types of cancers. It describes the initial therapeutic and pharmacological classes of the repurposed drugs and their new applications and mechanisms of action in cancer treatment. The review reports on drugs from various pharmacological classes that have been successfully repurposed for cancer treatment, including approved ones and those in clinical trials and preclinical development. It stratifies drugs based on their anticancer repurpose as multi-type, type-specific, and mechanism-directed, and according to their pharmacological classes. The review also reflects on the future potential that drug repurposing has in the clinical development of novel anticancer therapies.
Collapse
Affiliation(s)
- Abdulaziz H. Al Khzem
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohamed S. Gomaa
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mansour S. Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Nada Tawfeeq
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohammad Sarafroz
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Shareefa M. Alonaizi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Alhassan Al Faran
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Laela Ahmed Alrumaihi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Fatimah Ahmed Alansari
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Abdullah Abbas Alghamdi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| |
Collapse
|
8
|
Yin Y, Li Y, Ma B, Ren C, Zhao S, Li J, Gong Y, Yang H, Li J. Mitochondrial-Derived Peptide MOTS-c Suppresses Ovarian Cancer Progression by Attenuating USP7-Mediated LARS1 Deubiquitination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405620. [PMID: 39321430 DOI: 10.1002/advs.202405620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/13/2024] [Indexed: 09/27/2024]
Abstract
Mitochondrial-nuclear communication plays a vital role in maintaining cellular homeostasis. MOTS-c, a short peptide derived from the 12S rRNA of mitochondrial DNA, has been suggested as a retrograde mitochondrial signal. Although recent clinical studies have suggested a possible link between MOTS-c and human cancer, the role of MOTS-c in tumorigenesis has yet to be investigated. Here, MOTS-c levels are found to be reduced in both serum and tumor tissues from ovarian cancer (OC) patients, which are associated with poor patients' prognosis. Exogenous MOTS-c inhibits the proliferation, migration and invasion of OC cells, and induces cell cycle arrest and apoptosis. Mechanistically, MOTS-c interacts with LARS1 and promotes its ubiquitination and proteasomal degradation. In addition, USP7 was identified as a deubiquitinase of LARS1, and MOTS-c can attenuates USP7-mediated LARS1 deubiquitination by competing with USP7 for binding to LARS1. Besides, LARS1 was found to be increased and play an important oncogenic function in OC. More importantly, MOTS-c displays a marked anti-tumor effect on OC growth without systemic toxicity in vivo. In conclusion, this study reveals a crucial role of MOTS-c in OC and provides a possibility for MOTS-c as a therapeutic target for the treatment of this manlignacy.
Collapse
Affiliation(s)
- Yadong Yin
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yujie Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Boyi Ma
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Chenlu Ren
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shuhua Zhao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jia Li
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yun Gong
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jibin Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Department of Physiology and Pathophysiology, Air Force Medical University, Xi'an, 710032, China
| |
Collapse
|
9
|
Xiao Y, Pang N, Ma S, Gao M, Yang L. Effect of Nicotinamide Riboside Against the Exhaustion of CD8 + T Cells via Alleviating Mitochondrial Dysfunction. Nutrients 2024; 16:3577. [PMID: 39519411 PMCID: PMC11547570 DOI: 10.3390/nu16213577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Targeting mitochondria and protecting the mitochondrial function of CD8+ T cells are crucial for enhancing the clinical efficacy of cancer immunotherapy. Objectives: In this study, our objective was to investigate the potential of nicotinamide riboside (NR) in preserving the mitochondrial function of CD8+ T cells and mitigating their exhaustion. Methods: We established two in vitro models to induce CD8+ T cell exhaustion either by tumor cell-conditioned medium (TCM) or by continuous stimulation with OVA(257-264) peptide. CD8+ T cells were treated in the absence/presence of NR. Results: Our findings demonstrated that NR supplementation effectively inhibited CD8+ T cell exhaustion and preserved mitochondrial function in both models. Moreover, apoptosis of CD8+ T cells was reduced after NR treatment. Western blot data indicated that NR treatment upregulated Silent information regulator 1 (SirT1) expression. Further inhibition of Sirt1 activity using EX527 uncovered that the inhibitory effect of NR on CD8+ T cell exhaustion and its protective effect on mitochondria were attenuated. Conclusions: In conclusion, our results indicate that NR supplementation attenuates CD8+ T cell exhaustion, and its underlying mechanism is associated with increased mitochondrial function regulated by the SirT1 pathway. Our research provides evidence that NR may assist in enhancing the clinical efficacy of immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Lili Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition, and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
10
|
Salimi A, Asgari B, Khezri S, Pourgholi M, Haddadi S. Hesperidin as a bioactive compound in citrus fruits reduces N-ethyl-N-nitrosourea-induced mortality and toxicity in mice: as a model for chronic lymphocytic leukemia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03531-8. [PMID: 39400715 DOI: 10.1007/s00210-024-03531-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
The current study is aimed at determining the preventive effects of hesperidin against death, weight changes, cellular damage, and oxidative stress in mice induced by n-ethyl-n-nitrosourea as a chronic lymphocytic leukemia (CLL) model. Female mice were pretreated with hesperidin (20 mg/kg, intraperitoneally, daily for 30 days). Next, the animals received a single intraperitoneal injection of 80 mg/kg ENU on the 30th. Changes in weight and mortality were monitored for 120 days, and then the animals were sacrificed and parameters such as reactive oxygen species (ROS), mitochondrial dysfunction, lysosomal membrane integrity, oxidized/reduced glutathione (GSH/GSSG), and malondialdehyde (MDA) were analyzed in isolated lymphocytes. Hesperidin significantly increases the survival of mice up to 86% and delay in death time and prevents weight changes after exposure to ENU. Also, hesperidin improved cellular toxicity parameters such as ROS formation, MMP collapse, lysosomal membrane destabilization, and lipid peroxidation in isolated lymphocytes. These results promisingly showed that pretreatment with hesperidin increases delay in death time and reduces mortality cellular toxicities consistent with the carcinogenicity of alkylating compounds. This study confirms that the consumption of hesperidin and citrus most likely inhibits alkylating agents-induced carcinogenicity and toxicity.
Collapse
Affiliation(s)
- Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.
- Traditional Medicine and Hydrotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Bahare Asgari
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saleh Khezri
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahshad Pourgholi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shadi Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
11
|
Ali-El-Dein B, Abdelgawad M, Tarek M, Abdel-Rahim M, Elkady ME, Saleh HH, Zakaria MM, Tarabay HH, Laymon M, Mosbah A, Stenzl A. Bladder cancer associated with elevated heavy metals: Investigation of probable carcinogenic pathways through mitochondrial dysfunction, oxidative stress and mitogen-activated protein kinase. Urol Oncol 2024:S1078-1439(24)00650-1. [PMID: 39379209 DOI: 10.1016/j.urolonc.2024.09.009] [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: 08/01/2024] [Revised: 09/01/2024] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
OBJECTIVE Carcinogenic mechanisms of heavy metals/ trace elements (HMTE) in bladder cancer (BC) are exactly unknown. Mitochondrial dysfunction (MD), oxidative stress (OS), and mitogen-activated protein kinases (MAPK) are probable carcinogenic mechanisms. The purpose is to investigate probable carcinogenic pathways of HMTE in BC using six MD genes, seven OS markers, and p38-MAPK. METHODS Study included 125 BC/radical cystectomy (RC) patients between October 2020 and October 2022, and 72 controls. Exclusion criteria included previous neoplasm, chemo- or radiotherapy. Two samples (cancer/noncancer) were taken from RC specimens. Tissues/plasma/urine cadmium (Cd), lead (Pb), cobalt (Co), nickel (Ni), strontium (Sr), aluminium (Al), zinc (Zn), boron (B) were measured by ICP-OES. Tissue MD genes (mt-CO3, mt-CYB, mt-ATP 6, mt-ATP8, mt-CO1, mt-ND1), and serum OS markers (8-OHdG, MDA, 3-NT, AGEs, AOPP, ROS, SOD2), p38-MAPK were assessed by RT-PCR, and ELISA, respectively. RESULTS BC and adjacent tissue showed higher (Al, Co, Pb, Ni, Zn, Cd,Sr), lower B concentrations, compared to controls. High tissue concentrations (Cd, Co, Pb, Ni, Sr) were associated with higher MD genes, OS, MAPK and lower SOD2 levels. The same differences were greater in 41 patients with concomitant elevation of two or more HMTE. Noninclusion of BC-related oncogenes (e.g. RAS) is a limitation. CONCLUSIONS Evidence suggests that high BC tissue (Cd, Co, Pb, Ni, Si) concentrations are associated with over-expressed MD genes, OS, p38-MAPK and low SOD2. These findings provide important understanding keys of probable carcinogenic pathways in BC associated with HMTE. So, efforts should be performed to minimize and counteract exposure to toxic HMTE.
Collapse
Affiliation(s)
- Bedeir Ali-El-Dein
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | | | - Mohamed Tarek
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mona Abdel-Rahim
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Manar E Elkady
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hazem H Saleh
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mahmoud M Zakaria
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Heba H Tarabay
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mahmoud Laymon
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Mosbah
- Urology and Nephrology Center, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Arnolf Stenzl
- Department of Urology, University of Tuebingen Medical School, Tuebingen, Germany
| |
Collapse
|
12
|
Zhang Y, Gong Y, Liang Z, Wu W, Chen J, Li Y, Chen R, Mei J, Huang Z, Sun J. Mitochondria- and endoplasmic reticulum-localizing iridium(III) complexes induce immunogenic cell death of 143B cells. J Inorg Biochem 2024; 259:112655. [PMID: 38943844 DOI: 10.1016/j.jinorgbio.2024.112655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Recent breakthroughs in cancer immunology have propelled immunotherapy to the forefront of cancer research as a promising treatment approach that harnesses the body's immune system to effectively identify and eliminate cancer cells. In this study, three novel cyclometalated Ir(III) complexes, Ir1, Ir2, and Ir3, were designed, synthesized, and assessed in vitro for cytotoxic activity against several tumor-derived cell lines. Among these, Ir1 exhibited the highest cytotoxic activity, with an IC50 value of 0.4 ± 0.1 μM showcasing its significant anticancer potential. Detailed mechanistic analysis revealed that co-incubation of Ir1 with 143B cells led to Ir1 accumulation within mitochondria and the endoplasmic reticulum (ER). Furthermore, Ir1 induced G0/G1 phase cell cycle arrest, while also diminishing mitochondrial membrane potential, disrupting mitochondrial function, and triggering ER stress. Intriguingly, in mice the Ir1-induced ER stress response disrupted calcium homeostasis to thereby trigger immunogenic cell death (ICD), which subsequently activated the host antitumor immune response while concurrently dampening the in vivo tumor-induced inflammatory response.
Collapse
Affiliation(s)
- Yuqing Zhang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yao Gong
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhijun Liang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wei Wu
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxi Chen
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Yuling Li
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Rui Chen
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jun Mei
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| | - Jing Sun
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
13
|
Zhang F, Guo J, Yu S, Zheng Y, Duan M, Zhao L, Wang Y, Yang Z, Jiang X. Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment. Cancer Commun (Lond) 2024; 44:929-966. [PMID: 38997794 PMCID: PMC11492308 DOI: 10.1002/cac2.12591] [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: 11/26/2023] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024] Open
Abstract
The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.
Collapse
Affiliation(s)
- Fusheng Zhang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
- Department of Hepatobiliary and Pancreatic SurgeryPeking University First HospitalBeijingP. R. China
| | - Junchen Guo
- Department of RadiologyThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Shengmiao Yu
- Outpatient DepartmentThe Fourth Affiliated HospitalChina Medical UniversityShenyangLiaoningP. R. China
| | - Youwei Zheng
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Meiqi Duan
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Liang Zhao
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Yihan Wang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Zhi Yang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Xiaofeng Jiang
- Department of General SurgeryThe Fourth Affiliated Hospital of China Medical UniversityShenyangLiaoningP. R. China
| |
Collapse
|
14
|
El-Tanani M, Rabbani SA, El-Tanani Y, Matalka II. Metabolic vulnerabilities in cancer: A new therapeutic strategy. Crit Rev Oncol Hematol 2024; 201:104438. [PMID: 38977145 DOI: 10.1016/j.critrevonc.2024.104438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Cancer metabolism is now a key area for therapeutic intervention, targeting unique metabolic reprogramming crucial for tumor growth and survival. This article reviews the therapeutic potential of addressing metabolic vulnerabilities through glycolysis and glutaminase inhibitors, which disrupt cancer cell metabolism. Challenges such as tumor heterogeneity and adaptive resistance are discussed, with strategies including personalized medicine and predictive biomarkers to enhance treatment efficacy. Additionally, integrating diet and lifestyle changes with metabolic targeting underscores a holistic approach to improving therapy outcomes. The article also examines the benefits of incorporating these strategies into standard care, highlighting the potential for more tailored, safer treatments. In conclusion, exploiting metabolic vulnerabilities promises a new era in oncology, positioning metabolic targeting at the forefront of personalized cancer therapy and transforming patient care.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Yahia El-Tanani
- Medical School, St George's University of London, Cranmer Terrace, Tooting, London, UK
| | - Ismail I Matalka
- RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Department of Pathology and Microbiology, Medicine, Jordan University of Science and Technology, Irbid, Jordan.
| |
Collapse
|
15
|
Lee HT, Lin CS, Liu CY, Chen P, Tsai CY, Wei YH. Mitochondrial Plasticity and Glucose Metabolic Alterations in Human Cancer under Oxidative Stress-From Viewpoints of Chronic Inflammation and Neutrophil Extracellular Traps (NETs). Int J Mol Sci 2024; 25:9458. [PMID: 39273403 PMCID: PMC11395599 DOI: 10.3390/ijms25179458] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress elicited by reactive oxygen species (ROS) and chronic inflammation are involved both in deterring and the generation/progression of human cancers. Exogenous ROS can injure mitochondria and induce them to generate more endogenous mitochondrial ROS to further perpetuate the deteriorating condition in the affected cells. Dysfunction of these cancer mitochondria may possibly be offset by the Warburg effect, which is characterized by amplified glycolysis and metabolic reprogramming. ROS from neutrophil extracellular traps (NETs) are an essential element for neutrophils to defend against invading pathogens or to kill cancer cells. A chronic inflammation typically includes consecutive NET activation and tissue damage, as well as tissue repair, and together with NETs, ROS would participate in both the destruction and progression of cancers. This review discusses human mitochondrial plasticity and the glucose metabolic reprogramming of cancer cells confronting oxidative stress by the means of chronic inflammation and neutrophil extracellular traps (NETs).
Collapse
Affiliation(s)
- Hui-Ting Lee
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Chen-Sung Lin
- Division of Thoracic Surgery, Department of Surgery, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for General Education, Kainan University, Taoyuan City 338, Taiwan
| | - Chao-Yu Liu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Po Chen
- Cancer Free Biotech, Taipei 114, Taiwan
| | - Chang-Youh Tsai
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Clinical Trial Center, Division of Immunology & Rheumatology, Fu Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Faculty of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
16
|
Ba Q, Wang X, Lu Y. Establishment of a prognostic model for pancreatic cancer based on mitochondrial metabolism related genes. Discov Oncol 2024; 15:376. [PMID: 39196457 PMCID: PMC11358576 DOI: 10.1007/s12672-024-01255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
AIM Pancreatic ductal adenocarcinoma (PAAD) is recognized as an exceptionally aggressive cancer that both highly lethal and unfavorable prognosis. The mitochondrial metabolism pathway is intimately involved in oncogenesis and tumor progression, however, much remains unknown in this area. In this study, the bioinformatic tools have been used to construct a prognostic model with mitochondrial metabolism-related genes (MMRGs) to evaluate the survival, immune status, mutation profile, and drug sensitivity of PAAD patients. METHOD Univariate Cox regression and LASSO regression were used to screen the differentially expressed genes (DEGs), and multivariate Cox regression was used to develop the risk model. Kaplan-Meier estimator was employed to identify MMRGs signatures associated with overall survival (OS). ROC curves were utilized to evaluate the model's performance. Maftools, immunedeconv and CIBERSORT R packages were applied to analyze the gene mutation profiles and immune status. The corresponding sensitivity to pharmaceutical agents was assessed using oncoPredict R packages. RESULTS A prognostic model with five MMRGs was developed, which defined the patients as high-risk showed lower survival rates. There was good consistency among individuals categorized as high-risk, showing elevated rates of genetic alterations, particularly in the TP53 and KRAS genes. Furthermore, these patients exhibited increased levels of immunosuppression, characterized by an increased presence of macrophages, neutrophils, Th2 cells, and regulatory T cells. Additionally, high-risk patients showed increased sensitivity to Sabutoclax and Venetoclax. CONCLUSION By utilizing a gene signature associated with mitochondrial metabolism, a prognostic model has been established which could be a highly efficient method for predicting the outcomes of PAAD patients.
Collapse
Affiliation(s)
- Qinwen Ba
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Bednarska-Szczepaniak K, Hałagan K, Szwed M, Przelazły E, Leśnikowski ZJ. Quantum Chemical and Biological Insights into Redox Activity of Metallacarborane Complexes in Cancer Cells. J Chem Inf Model 2024; 64:6521-6541. [PMID: 39140958 DOI: 10.1021/acs.jcim.4c00394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
A relationship between the electronic properties of metal ions in metallacarboranes and their ability to modulate mitochondrial oxidase activity and membrane hyperpolarization in cancer cells was demonstrated. Quantum chemistry methods, including DFT and molecular dynamics simulations, were used to understand the oxidized and reduced forms of metallacarboranes and their intramolecular rotatory behavior. According to the low-spin assumption for metal ions, the intramolecular oscillations of cluster ligands in metallacarboranes are significantly influenced by the type of metal and correspond to the cellular uptake of these complexes in vitro. In particular, the low-spin iron compound may be a new xenogeneic booster of redox homeostasis in cancer cells resistant to cisplatin, which induces metabolic 'exhaustion' of cancer cells and their death.
Collapse
Affiliation(s)
- Katarzyna Bednarska-Szczepaniak
- Laboratory of Medicinal Chemistry, Polish Academy of Sciences, Institute of Medical Biology, 106 Lodowa, 92-232 Lodz, Poland
| | - Krzysztof Hałagan
- Department of Molecular Physics, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Lodz, Poland
| | - Marzena Szwed
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Institute of Biophysics, Pomorska 141/143, 90-236 Lodz, Poland
| | - Ewelina Przelazły
- Laboratory of Medicinal Chemistry, Polish Academy of Sciences, Institute of Medical Biology, 106 Lodowa, 92-232 Lodz, Poland
| | - Zbigniew J Leśnikowski
- Laboratory of Medicinal Chemistry, Polish Academy of Sciences, Institute of Medical Biology, 106 Lodowa, 92-232 Lodz, Poland
| |
Collapse
|
18
|
Arast Y, Sabbaghi M, Kamranfar F, Heidari F, Fazli Nejad SM, Hosseinabadi T, Pourahmad J. Selective cytotoxicity of standardised n-hexane extract of black soldier flies' larvae on cancerous skin cells mitochondria isolated from rat model of melanoma. Cutan Ocul Toxicol 2024:1-8. [PMID: 39115252 DOI: 10.1080/15569527.2024.2389193] [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: 12/27/2023] [Revised: 06/30/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
INTRODUCTION Melanoma is known as a highly lethal cancer. In melanoma cells, apoptosis signalling which relies heavily on the acute activity of mitochondria and reactive oxygen species (ROS) formation is suppressed. Our previous studies on natural compounds on melanoma suggested that mitochondria are a potential target for the melanoma treatment by selective cytotoxic effect of them. The black soldier fly is an important environmental protectant insect that based on recent studies induces apoptosis in liver and colorectal carcinoma cells through the activation of caspase 3, 8, and 9 and ultimately inhibits the growth of cancer cells. PURPOSE This study was designed to evaluate the selective apoptotic effect of the n-hexane BSFL extract (BSFLE) on skin mitochondria. MATERIALS AND METHODS The mitochondria isolated from melanoma cells were treated with various concentrations (1500, 3000, and 6000 µg/ml) of n-hexane BSFLE Then MTT viability assay, ROS determination, Mitochondrial Membrane Potential (MMP), mitochondrial swelling, cytochrome c release determination, and % apoptosis were performed. RESULTS MTT assay showed that different concentrations of n-hexane BSFLE significantly (P < 0.05) decreased the SDH activity in cancerous skin mitochondria with the IC50. The ROS production and mitochondrial swelling results also showed that all concentrations of BSFL extracts significantly increased. MMP decline and the release of cytochrome c in cancer groups mitochondria. BSFLE increased apoptosis on melanoma cells. DISCUSSION AND CONCLUSION It is suggested that n-hexane BSFLE compounds selectively induce a cascade of proapoptotic events that are probably defective in cancer cells. Most of these compounds target the mitochondrial transient pore caused by disruption of the mitochondrial respiratory chain. These events lead to disruption of the temporary permeability of mitochondria, swelling of mitochondria and finally the formation of apoptosome in the cytosol.
Collapse
Affiliation(s)
- Yalda Arast
- Research Center of Environmental Pollutants, Qom University of Medical Sciences, Qom, Iran
| | - Mahya Sabbaghi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Tehran, Iran
| | - Farzane Kamranfar
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Tehran, Iran
| | - Fatemeh Heidari
- Department of Anatomical Sciences, School of Medicine, Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | | | - Tahereh Hosseinabadi
- Department of Pharmacognosy and Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Tehran, Iran
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences Tehran, Tehran, Iran
| |
Collapse
|
19
|
Fu J, Li D, Zhang L, Maghsoudloo M, Cheng J, Fu J. Comprehensive analysis, diagnosis, prognosis, and cordycepin (CD) regulations for GSDME expressions in pan-cancers. Cancer Cell Int 2024; 24:279. [PMID: 39118110 PMCID: PMC11312966 DOI: 10.1186/s12935-024-03467-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
The Gasdermin E gene (GSDME) plays roles in deafness and cancers. However, the roles and mechanisms in cancers are complex, and the same gene exhibits different mechanisms and actions in different types of cancers. Online databases, such as GEPIA2, cBioPortal, and DNMIVD, were used to comprehensively analyze GSDME profiles, DNA methylations, mutations, diagnosis, and prognosis in patients with tumor tissues and matched healthy tissues. Western blotting and RT-PCR were used to monitor the regulation of GSDME by Cordycepin (CD) in cancer cell lines. We revealed that GSDME expression is significantly upregulated in eight cancers (ACC, DLBC, GBM, HNSC, LGG, PAAD, SKCM, and THYM) and significantly downregulated in seven cancers (COAD, KICH, LAML, OV, READ, UCES, and UCS). The overall survival was longer only in ACC, but shorter in four cancers, including COAD, KIRC, LIHC, and STAD, when GSDME was highly expressed in cancers compared with the corresponding normal tissues. Moreover, the high expression of GSDME was negatively correlated with the poor prognosis of ACC, while the low expression of GSDME was negatively correlated with the poor prognosis of COAD, suggesting that GSDME might serve as a good prognostic factor in these two cancer types. Accordingly, results indicated that the DNA methylations of those 7 CpG sites constitute a potentially effective signature to distinguish different tumors from adjacent healthy tissues. Gene mutations for GSDME were frequently observed in a variety of tumors, with UCES having the highest frequency. Moreover, CD treatment inhibited GSDME expression in different cancer cell lines, while overexpression of GSDME promoted cell migration and invasion. Thus, we have systematically and successfully clarified the GSDME expression profiles, diagnostic values, and prognostic values in pan-cancers. Targeting GSDME with CD implies therapeutic significance and a mechanism for antitumor roles in some types of cancers via increasing the sensitivity of chemotherapy. Altogether, our study may provide a strategy and biomarker for clinical diagnosis, prognostics, and treatment of cancers by targeting GSDME.
Collapse
Affiliation(s)
- Jiewen Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Dabing Li
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Lianmei Zhang
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China
- Department of Pathology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu Province, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China.
| | - Jingliang Cheng
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China.
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan Province, P R China.
| |
Collapse
|
20
|
Yang T, Liu Z, Zhang T, Liu Y. Hybrid nano-stimulator for specific amplification of oxidative stress and precise tumour treatment. J Drug Target 2024; 32:756-769. [PMID: 38832845 DOI: 10.1080/1061186x.2024.2349112] [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: 12/01/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND The use of reactive oxygen species (ROS) to target cancer cells has become a hot topic in tumor therapy. PURPOSE Although ROS has strong cytotoxicity against tumor cells, the key issue currently is how to generate a large amount of ROS within tumor cells. METHODS Organic/inorganic hybrid nanoreactor materials combine the advantages of organic and inorganic components and can amplify cancer treatment by increasing targeting and material self-action. The multifunctional organic / inorganic hybrid nanoreactor is helpful to overcome the shortcomings of current reactive oxygen species in cancer treatment. It can realize the combination of in situ dynamic therapy and immunotherapy strategies, and has a synergistic anti-tumor effect. RESULTS This paper reviews the research progress of organic/inorganic hybrid nanoreactor materials using tumor components to amplify reactive oxygen species for cancer treatment. The article reviews the tumor treatment strategies of nanohybrids from the perspectives of cancer cells, immune cells, tumor microenvironment, as well as 3D printing and electrospinning techniques, which are different from traditional nanomaterial technologies, and will arouse interest among scientists in tumor therapy and nanomedicine.
Collapse
Affiliation(s)
- Ting Yang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Zihan Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tong Zhang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Yinchuan, China
| |
Collapse
|
21
|
Zang Y, Wang A, Zhang J, Xia M, Jiang Z, Jia B, Lu C, Chen C, Wang S, Zhang Y, Wang C, Cao X, Niu Z, He C, Bai X, Tian S, Zhai G, Cao H, Chen Y, Zhang K. Hypoxia promotes histone H3K9 lactylation to enhance LAMC2 transcription in esophageal squamous cell carcinoma. iScience 2024; 27:110188. [PMID: 38989468 PMCID: PMC11233973 DOI: 10.1016/j.isci.2024.110188] [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: 02/22/2024] [Revised: 05/07/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Hypoxia promotes tumorigenesis and lactate accumulation in esophageal squamous cell carcinoma (ESCC). Lactate can induce histone lysine lactylation (Kla, a recently identified histone marks) to regulate transcription. However, the functional consequence of histone Kla under hypoxia in ESCC remains to be explored. Here, we reveal that hypoxia facilitates histone H3K9la to enhance LAMC2 transcription for proliferation of ESCC. We found that global level of Kla was elevated under hypoxia, and thus identified the landscape of histone Kla in ESCC by quantitative proteomics. Furthermore, we show a significant increase of H3K9la level induced by hypoxia. Next, MNase ChIP-seq and RNA-seq analysis suggest that H3K9la is enriched at the promoter of cell junction genes. Finally, we demonstrate that the histone H3K9la facilitates the expression of LAMC2 for ESCC invasion by in vivo and in vitro experiments. Briefly, our study reveals a vital role of histone Kla triggered by hypoxia in cancer.
Collapse
Affiliation(s)
- Yong Zang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Aiyuan Wang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jianji Zhang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Mingxin Xia
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zixin Jiang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Bona Jia
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Congcong Lu
- Frontier Center for Cell Response, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chen Chen
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Siyu Wang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yingao Zhang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Chen Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Xinyi Cao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ziping Niu
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Chaoran He
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xue Bai
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Shanshan Tian
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Guijin Zhai
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yupeng Chen
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Kai Zhang
- 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, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
22
|
Cela O, Scrima R, Pacelli C, Rosiello M, Piccoli C, Capitanio N. Autonomous Oscillatory Mitochondrial Respiratory Activity: Results of a Systematic Analysis Show Heterogeneity in Different In Vitro-Synchronized Cancer Cells. Int J Mol Sci 2024; 25:7797. [PMID: 39063035 PMCID: PMC11276763 DOI: 10.3390/ijms25147797] [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: 06/18/2024] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Circadian oscillations of several physiological and behavioral processes are an established process in all the organisms anticipating the geophysical changes recurring during the day. The time-keeping mechanism is controlled by a transcription translation feedback loop involving a set of well-characterized transcription factors. The synchronization of cells, controlled at the organismal level by a brain central clock, can be mimicked in vitro, pointing to the notion that all the cells are endowed with an autonomous time-keeping system. Metabolism undergoes circadian control, including the mitochondrial terminal catabolic pathways, culminating under aerobic conditions in the electron transfer to oxygen through the respiratory chain coupled to the ATP synthesis according to the oxidative phosphorylation chemiosmotic mechanism. In this study, we expanded upon previous isolated observations by utilizing multiple cell types, employing various synchronization protocols and different methodologies to measure mitochondrial oxygen consumption rates under conditions simulating various metabolic stressors. The results obtained clearly demonstrate that mitochondrial respiratory activity undergoes rhythmic oscillations in all tested cell types, regardless of their individual respiratory proficiency, indicating a phenomenon that can be generalized. However, notably, while primary cell types exhibited similar rhythmic respiratory profiles, cancer-derived cell lines displayed highly heterogeneous rhythmic changes. This observation confirms on the one hand the dysregulation of the circadian control of the oxidative metabolism observed in cancer, likely contributing to its development, and on the other hand underscores the necessity of personalized chronotherapy, which necessitates a detailed characterization of the cancer chronotype.
Collapse
Affiliation(s)
- Olga Cela
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (M.R.); (C.P.); (N.C.)
| | - Rosella Scrima
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (C.P.); (M.R.); (C.P.); (N.C.)
| | | | | | | | | |
Collapse
|
23
|
Hamwi MN, Elsayed E, Dabash H, Abuawad A, Aweer NA, Al Zeir F, Pedersen S, Al-Mansoori L, Burgon PG. MLIP and Its Potential Influence on Key Oncogenic Pathways. Cells 2024; 13:1109. [PMID: 38994962 PMCID: PMC11240681 DOI: 10.3390/cells13131109] [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: 01/26/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
Muscle-enriched A-type lamin-interacting protein (MLIP) is an emerging protein involved in cellular homeostasis and stress adaptation. Eukaryotic cells regulate various cellular processes, including metabolism, DNA repair, and cell cycle progression, to maintain cellular homeostasis. Disruptions in this homeostasis can lead to diseases such as cancer, characterized by uncontrolled cell growth and division. This review aims to explore for the first time the unique role MLIP may play in cancer development and progression, given its interactions with the PI3K/Akt/mTOR pathway, p53, MAPK9, and FOXO transcription factors, all critical regulators of cellular homeostasis and tumor suppression. We discuss the current understanding of MLIP's involvement in pro-survival pathways and its potential implications in cancer cells' metabolic remodeling and dysregulated homeostasis. Additionally, we examine the potential of MLIP as a novel therapeutic target for cancer treatment. This review aims to shed light on MLIP's potential impact on cancer biology and contribute to developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mahmoud N Hamwi
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Engy Elsayed
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Hanan Dabash
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Amani Abuawad
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Noor A Aweer
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Faissal Al Zeir
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Shona Pedersen
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Layla Al-Mansoori
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Patrick G Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
24
|
Wang C, Tan J, Jin Y, Li Z, Yang J, Jia Y, Xia Y, Gong B, Dong Q, Zhao Q. A mitochondria-related genes associated neuroblastoma signature - based on bulk and single-cell transcriptome sequencing data analysis, and experimental validation. Front Immunol 2024; 15:1415736. [PMID: 38962012 PMCID: PMC11220120 DOI: 10.3389/fimmu.2024.1415736] [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: 04/11/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Background Neuroblastoma (NB), characterized by its marked heterogeneity, is the most common extracranial solid tumor in children. The status and functionality of mitochondria are crucial in regulating NB cell behavior. While the significance of mitochondria-related genes (MRGs) in NB is still missing in key knowledge. Materials and methods This study leverages consensus clustering and machine learning algorithms to construct and validate an MRGs-related signature in NB. Single-cell data analysis and experimental validation were employed to characterize the pivotal role of FEN1 within NB cells. Results MRGs facilitated the classification of NB patients into 2 distinct clusters with considerable differences. The constructed MRGs-related signature and its quantitative indicators, mtScore and mtRisk, effectively characterize the MRGs-related patient clusters. Notably, the MRGs-related signature outperformed MYCN in predicting NB patient prognosis and was adept at representing the tumor microenvironment (TME), tumor cell stemness, and sensitivity to the chemotherapeutic agents Cisplatin, Topotecan, and Irinotecan. FEN1, identified as the most contributory gene within the MRGs-related signature, was found to play a crucial role in the communication between NB cells and the TME, and in the developmental trajectory of NB cells. Experimental validations confirmed FEN1's significant influence on NB cell proliferation, apoptosis, cell cycle, and invasiveness. Conclusion The MRGs-related signature developed in this study offers a novel predictive tool for assessing NB patient prognosis, immune infiltration, stemness, and chemotherapeutic sensitivity. Our findings unveil the critical function of FEN1 in NB, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxiong Tan
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yan Jin
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Zongyang Li
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Jiaxing Yang
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yubin Jia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yuren Xia
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Baocheng Gong
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiuping Dong
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Qiang Zhao
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
25
|
Wang Y, He X, Cheng N, Huang K. Unveiling the Nutritional Veil of Sulforaphane: With a Major Focus on Glucose Homeostasis Modulation. Nutrients 2024; 16:1877. [PMID: 38931232 PMCID: PMC11206418 DOI: 10.3390/nu16121877] [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: 05/23/2024] [Revised: 06/09/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Abnormal glucose homeostasis is associated with metabolic syndromes including cardiovascular diseases, hypertension, type 2 diabetes mellitus, and obesity, highlighting the significance of maintaining a balanced glucose level for optimal biological function. This highlights the importance of maintaining normal glucose levels for proper biological functioning. Sulforaphane (SFN), the primary bioactive compound in broccoli from the Cruciferae or Brassicaceae family, has been shown to enhance glucose homeostasis effectively while exhibiting low cytotoxicity. This paper assesses the impact of SFN on glucose homeostasis in vitro, in vivo, and human trials, as well as the molecular mechanisms that drive its regulatory effects. New strategies have been proposed to enhance the bioavailability and targeted delivery of SFN in order to overcome inherent instability. The manuscript also covers the safety evaluations of SFN that have been documented for its production and utilization. Hence, a deeper understanding of the favorable influence and mechanism of SFN on glucose homeostasis, coupled with the fact that SFN is abundant in the human daily diet, may ultimately offer theoretical evidence to support its potential use in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Yanan Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Nan Cheng
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| | - Kunlun Huang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.W.); (X.H.); (N.C.)
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs of the P.R. China, Beijing 100083, China
| |
Collapse
|
26
|
Xu Z, Chen X, Zhou H, Sun L, Bai R, Yu W, Yang J, Liu H. The clinical significance of mitochondrial calcium uniporter in gastric cancer patients and its preliminary exploration of the impact on mitochondrial function and metabolism. Front Oncol 2024; 14:1355559. [PMID: 38737905 PMCID: PMC11082321 DOI: 10.3389/fonc.2024.1355559] [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: 12/14/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Objective The objective of this study is to elucidate the influence of MCU on the clinical pathological features of GC patients, to investigate the function and mechanism of the mitochondrial calcium uptake transporter MCU in the initiation and progression of GC, and to explore its impact on the metabolic pathways and biosynthesis of mitochondria. The ultimate goal is to identify novel targets and strategies for the clinical management of GC patients. Methods Tumor and adjacent tissue specimens were obtained from 205 patients with gastric cancer, and immunohistochemical tests were performed to assess the expression of MCU and its correlation with clinical pathological characteristics and prognosis. Data from TCGA, GTEx and GEO databases were retrieved for gastric cancer patients, and bioinformatics analysis was utilized to investigate the association between MCU expression and clinical pathological features. Furthermore, we conducted an in-depth analysis of the role of MCU in GC patients. We investigated the correlation between MCU expression in GC and its impact on mitochondrial function, metabolism, biosynthesis, and immune cells. Additionally, we studied the proteins or molecules that interact with MCU. Results Our research revealed high expression of MCU in the GC tissues. This high expression was associated with poorer T and N staging, and indicated a worse disease-free survival period. MCU expression was positively correlated with mitochondrial function, mitochondrial metabolism, nucleotide, amino acid, and fatty acid synthesis metabolism, and negatively correlated with nicotinate and nicotinamide metabolism. Furthermore, the MCU also regulates the function of the mitochondrial oxidative respiratory chain. The MCU influences the immune cells of GC patients and regulates ROS generation, cell proliferation, apoptosis, and resistance to platinum-based drugs in gastric cancer cells. Conclusion High expression of MCU in GC indicates poorer clinical outcomes. The expression of the MCU are affected through impacts the function of mitochondria, energy metabolism, and cellular biosynthesis in gastric cancer cells, thereby influencing the growth and metastasis of gastric cancer cells. Therefore, the mitochondrial changes regulated by MCU could be a new focus for research and treatment of GC.
Collapse
Affiliation(s)
- Zipeng Xu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Department of General Surgery, Chang An Hospital, Xian, China
| | - Xia Chen
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Department of General Surgery, Chang An Hospital, Xian, China
| | - Haicun Zhou
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Department of General Surgery, Chang An Hospital, Xian, China
| | - Luming Sun
- Gansu Provincial Key Laboratory of Stem Cell and Gene Medicine, The 940th Hospital of Joint Lohistica Support force of Chinese People’s Liberation Army, Lanzhou, China
| | - Ruobing Bai
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Wenwen Yu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Department of General Surgery, Chang An Hospital, Xian, China
| | - Junhao Yang
- Gansu Provincial Key Laboratory of Stem Cell and Gene Medicine, The 940th Hospital of Joint Lohistica Support force of Chinese People’s Liberation Army, Lanzhou, China
| | - Hongbin Liu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Gansu Provincial Key Laboratory of Stem Cell and Gene Medicine, The 940th Hospital of Joint Lohistica Support force of Chinese People’s Liberation Army, Lanzhou, China
| |
Collapse
|
27
|
Zheng BX, Long W, Zheng W, Zeng Y, Guo XC, Chan KH, She MT, Leung ASL, Lu YJ, Wong WL. Mitochondria-Selective Dicationic Small-Molecule Ligand Targeting G-Quadruplex Structures for Human Colorectal Cancer Therapy. J Med Chem 2024; 67:6292-6312. [PMID: 38624086 DOI: 10.1021/acs.jmedchem.3c02240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Mitochondria are important drug targets for anticancer and other disease therapies. Certain human mitochondrial DNA sequences capable of forming G-quadruplex structures (G4s) are emerging drug targets of small molecules. Despite some mitochondria-selective ligands being reported for drug delivery against cancers, the ligand design is mostly limited to the triphenylphosphonium scaffold. The ligand designed with lipophilic small-sized scaffolds bearing multipositive charges targeting the unique feature of high mitochondrial membrane potential (MMP) is lacking and most mitochondria-selective ligands are not G4-targeting. Herein, we report a new small-sized dicationic lipophilic ligand to target MMP and mitochondrial DNA G4s to enhance drug delivery for anticancer. The ligand showed marked alteration of mitochondrial gene expression and substantial induction of ROS production, mitochondrial dysfunction, DNA damage, cellular senescence, and apoptosis. The ligand also exhibited high anticancer activity against HCT116 cancer cells (IC50, 3.4 μM) and high antitumor efficacy in the HCT116 tumor xenograft mouse model (∼70% tumor weight reduction).
Collapse
Affiliation(s)
- Bo-Xin Zheng
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Wei Long
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Wende Zheng
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| | - Yaoxun Zeng
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| | - Xiao-Chun Guo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Ka-Hin Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Meng-Ting She
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| | - Alan Siu-Lun Leung
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
| | - Yu-Jing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| |
Collapse
|
28
|
Tan X, Wang S, Xiao GY, Wu C, Liu X, Zhou B, Jiang Y, Duose DY, Xi Y, Wang J, Gupta K, Pataer A, Roth JA, Kim MP, Chen F, Creighton CJ, Russell WK, Kurie JM. Chromosomal 3q amplicon encodes essential regulators of secretory vesicles that drive secretory addiction in cancer. J Clin Invest 2024; 134:e176355. [PMID: 38662435 PMCID: PMC11178546 DOI: 10.1172/jci176355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/12/2024] [Indexed: 06/18/2024] Open
Abstract
Cancer cells exhibit heightened secretory states that drive tumor progression. Here, we identify a chromosome 3q amplicon that serves as a platform for secretory regulation in cancer. The 3q amplicon encodes multiple Golgi-resident proteins, including the scaffold Golgi integral membrane protein 4 (GOLIM4) and the ion channel ATPase Secretory Pathway Ca2+ Transporting 1 (ATP2C1). We show that GOLIM4 recruits ATP2C1 and Golgi phosphoprotein 3 (GOLPH3) to coordinate calcium-dependent cargo loading and Golgi membrane bending and vesicle scission. GOLIM4 depletion disrupts the protein complex, resulting in a secretory blockade that inhibits the progression of 3q-amplified malignancies. In addition to its role as a scaffold, GOLIM4 maintains intracellular manganese (Mn) homeostasis by binding excess Mn in the Golgi lumen, which initiates the routing of Mn-bound GOLIM4 to lysosomes for degradation. We show that Mn treatment inhibits the progression of multiple types of 3q-amplified malignancies by degrading GOLIM4, resulting in a secretory blockade that interrupts pro-survival autocrine loops and attenuates pro-metastatic processes in the tumor microenvironment. Potentially underlying the selective activity of Mn against 3q-amplified malignancies, ATP2C1 co-amplification increases Mn influx into the Golgi lumen, resulting in a more rapid degradation of GOLIM4. These findings show that functional cooperativity between co-amplified genes underlies heightened secretion and a targetable secretory addiction in 3q-amplified malignancies.
Collapse
Affiliation(s)
- Xiaochao Tan
- Department of Thoracic/Head and Neck Medical Oncology
| | - Shike Wang
- Department of Thoracic/Head and Neck Medical Oncology
| | - Guan-Yu Xiao
- Department of Thoracic/Head and Neck Medical Oncology
| | - Chao Wu
- Department of Thoracic/Head and Neck Medical Oncology
| | - Xin Liu
- Department of Thoracic/Head and Neck Medical Oncology
| | - Biyao Zhou
- Department of Thoracic/Head and Neck Medical Oncology
| | - Yu Jiang
- Department of Thoracic/Head and Neck Medical Oncology
| | | | - Yuanxin Xi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kunika Gupta
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Apar Pataer
- Department of Thoracic and Cardiovascular Surgery and
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery and
| | - Michael P. Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fengju Chen
- Department of Medicine and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Chad J. Creighton
- Department of Medicine and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
| | | |
Collapse
|
29
|
Wang S, El Jurdi N, Thyagarajan B, Prizment A, Blaes AH. Accelerated Aging in Cancer Survivors: Cellular Senescence, Frailty, and Possible Opportunities for Interventions. Int J Mol Sci 2024; 25:3319. [PMID: 38542292 PMCID: PMC10970400 DOI: 10.3390/ijms25063319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 06/02/2024] Open
Abstract
The population of cancer survivors has markedly increased due to the rapid improvements in cancer treatment. However, cancer survivors experience accelerated aging, which leads to chronic diseases and other age-related conditions, such as frailty. Those conditions may persist years after cancer diagnosis and treatment. Cellular senescence, a hallmark of aging, is one of the mechanisms that contribute to accelerated aging in cancer survivors. Several aging measures, including measures based on clinical markers and biomarkers, have been proposed to estimate the aging process, and some of them have shown associations with mortality and frailty in cancer survivors. Several anti-aging interventions, including lifestyle changes and anti-aging drugs, have been proposed. Future research, particularly in large-scale studies, is needed to determine the efficiency of these aging measures and anti-aging interventions before considering their application in clinics. This review focuses on the mechanisms of cellular senescence and accelerated aging in cancer survivors, assessment of the aging process using clinical markers and biomarkers, and the high prevalence of frailty in that population, as well as possible opportunities for anti-aging interventions. A deeper understanding of aging measures and anti-aging interventions in cancer survivors will contribute to the development of effective strategies to mitigate accelerated aging in cancer survivors and improve their quality of life.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Najla El Jurdi
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anna Prizment
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anne H. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
30
|
Chi H, Su L, Yan Y, Gu X, Su K, Li H, Yu L, Liu J, Wang J, Wu Q, Yang G. Illuminating the immunological landscape: mitochondrial gene defects in pancreatic cancer through a multiomics lens. Front Immunol 2024; 15:1375143. [PMID: 38510247 PMCID: PMC10953916 DOI: 10.3389/fimmu.2024.1375143] [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: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024] Open
Abstract
This comprehensive review delves into the complex interplay between mitochondrial gene defects and pancreatic cancer pathogenesis through a multiomics approach. By amalgamating data from genomic, transcriptomic, proteomic, and metabolomic studies, we dissected the mechanisms by which mitochondrial genetic variations dictate cancer progression. Emphasis has been placed on the roles of these genes in altering cellular metabolic processes, signal transduction pathways, and immune system interactions. We further explored how these findings could refine therapeutic interventions, with a particular focus on precision medicine applications. This analysis not only fills pivotal knowledge gaps about mitochondrial anomalies in pancreatic cancer but also paves the way for future investigations into personalized therapy options. This finding underscores the crucial nexus between mitochondrial genetics and oncological immunology, opening new avenues for targeted cancer treatment strategies.
Collapse
Affiliation(s)
- Hao Chi
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yalan Yan
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Xiang Gu
- Biology Department, Southern Methodist University, Dallas, TX, United States
| | - Ke Su
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lili Yu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Jue Wang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
| | - Guanhu Yang
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, and University Hospital, Macau University of Science and Technology, Macau, Macao SAR, China
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
31
|
Shaw P, Dwivedi SKD, Bhattacharya R, Mukherjee P, Rao G. VEGF signaling: Role in angiogenesis and beyond. Biochim Biophys Acta Rev Cancer 2024; 1879:189079. [PMID: 38280470 DOI: 10.1016/j.bbcan.2024.189079] [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: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 01/29/2024]
Abstract
Angiogenesis is a crucial process for tissue development, repair, and tumor survival. Vascular endothelial growth factor (VEGF) is a key driver secreted by cancer cells, promoting neovascularization. While VEGF's role in angiogenesis is well-documented, its influence on the other aspects in tumor microenvironemt is less discussed. This review elaborates on VEGF's impact on intercellular interactions within the tumor microenvironment, including how VEGF affects pericyte proliferation and migration and mediates interactions between tumor-associated macrophages and cancer cells, resulting in PDL-1-mediated immunosuppression and Nrf2-mediated epithelial-mesenchymal transition. The review discusses VEGF's involvement in intra-organelle crosstalk, tumor metabolism, stemness, and epithelial-mesenchymal transition. It also provides insights into current anti-VEGF therapies and their limitations in cancer treatment. Overall, this review aims to provide a thorough overview of the current state of knowledge concerning VEGF signaling and its impact, not only on angiogenesis but also on various other oncogenic processes.
Collapse
Affiliation(s)
- Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
32
|
Wang SF, Chang YL, Liu TY, Huang KH, Fang WL, Li AFY, Yeh TS, Hung GY, Lee HC. Mitochondrial dysfunction decreases cisplatin sensitivity in gastric cancer cells through upregulation of integrated stress response and mitokine GDF15. FEBS J 2024; 291:1131-1150. [PMID: 37935441 DOI: 10.1111/febs.16992] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/18/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Gastric neoplasm is a high-mortality cancer worldwide. Chemoresistance is the obstacle against gastric cancer treatment. Mitochondrial dysfunction has been observed to promote malignant progression. However, the underlying mechanism is still unclear. The mitokine growth differentiation factor 15 (GDF15) is a significant biomarker for mitochondrial disorder and is activated by the integrated stress response (ISR) pathway. The serum level of GDF15 was found to be correlated with the poor prognosis of gastric cancer patients. In this study, we found that high GDF15 protein expression might increase disease recurrence in adjuvant chemotherapy-treated gastric cancer patients. Moreover, treatment with mitochondrial inhibitors, especially oligomycin (a complex V inhibitor) and salubrinal (an ISR activator), respectively, was found to upregulate GDF15 and enhance cisplatin insensitivity of human gastric cancer cells. Mechanistically, it was found that the activating transcription factor 4-C/EBP homologous protein pathway has a crucial function in the heightened manifestation of GDF15. In addition, reactive oxygen species-activated general control nonderepressible 2 mediates the oligomycin-induced ISR, and upregulates GDF15. The GDF15-glial cell-derived neurotrophic factor family receptor a-like-ISR-cystine/glutamate transporter-enhanced glutathione production was found to be involved in cisplatin resistance. These results suggest that mitochondrial dysfunction might enhance cisplatin insensitivity through GDF15 upregulation, and targeting mitokine GDF15-ISR regulation might be a strategy against cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yuh-Lih Chang
- Department of Pharmacy, Taipei Veterans General Hospital, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yu Liu
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuo-Hung Huang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Wen-Liang Fang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan
- Department of Surgery, Gastric Cancer Medical Center, Taipei Veterans General Hospital, Taiwan
| | - Anna Fen-Yau Li
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Anatomical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Giun-Yi Hung
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Taipei Veterans General Hospital, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
33
|
Alves MG, Cabral LGS, Totti PGF, Azarias FR, Pomini KT, Rici REG, Laiso RAN, Maria DA. 2-Aminoethyl Dihydrogen Phosphate (2-AEH2P) Associated with Cell Metabolism-Modulating Drugs Presents a Synergistic and Pro-Apoptotic Effect in an In Vitro Model of the Ascitic Ehrlich Tumor. Biomedicines 2024; 12:109. [PMID: 38255214 PMCID: PMC10813795 DOI: 10.3390/biomedicines12010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
The progression and maintenance of cancer characteristics are associated with cellular components linked to the tumor and non-cellular components with pro-tumoral properties. Pharmacological association with antagonists of the cellular components of the tumor, such as anti- and pro-apoptotic drugs, represents a novel adjuvant strategy. In this study, the antiproliferative, pro-apoptotic, and pharmacological effects of the combination of monophosphoester 2-AEH2P with Simvastatin, Coenzyme Q10, the chemotherapeutic drug paclitaxel, and colony-stimulating factor (GM-CSF) were evaluated. Tests were conducted to determine cytotoxic activity using the MTT method, cell cycle phases, and fragmented DNA by flow cytometry, mitochondrial membrane potential, expression of cell markers Bcl2, TNF-α/DR-4, Cytochrome c, caspase 3, and P53, and analysis of drug combination profiles using Synergy Finder 2.0 Software. The results showed a synergistic effect among the combinations, compared to individual treatments with the monophosphoester and other drugs. In addition, there was modulation of marker expression, indicating a pro-apoptotic and immunomodulatory effect of 2-AEH2P. Pharmacological analysis revealed that tumor cells treated with GM-CSF + 2-AEH2P exhibited a synergistic effect, while groups of tumor cells treated with paclitaxel, Coenzyme Q10, and Simvastatin showed additive effects. Furthermore, treatment with the paclitaxel + 2-AEH2P combination (12 h) resulted in a significant reduction in mitochondrial membrane potential. Pharmacological combinations for normal cells did not exhibit deleterious effects compared to mammary carcinomatosis tumor (EAT) cells.
Collapse
Affiliation(s)
- Monique G. Alves
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| | - Laertty G. S. Cabral
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| | - Paulo G. F. Totti
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
| | - Felipe R. Azarias
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
| | - Karine T. Pomini
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
| | - Rose E. G. Rici
- Graduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marília (UNIMAR), Marília 17525-902, Brazil (R.E.G.R.)
- Graduate Program in Anatomy of Domestic and Wild Animals, College of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-220, Brazil
| | - Rosa A. N. Laiso
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
| | - Durvanei A. Maria
- Development and Innovation Laboratory, Butantan Institute, São Paulo 05359-900, Brazil; (L.G.S.C.); (F.R.A.)
- Graduate Program in Medical Sciences, College of Medicine, University of São Paulo, São Paulo 05508-220, Brazil
| |
Collapse
|
34
|
Li Z, Zhang W, Guo S, Qi G, Huang J, Gao J, Zhao J, Kang L, Li Q. A Review of Advances in Mitochondrial Research in Cancer. Cancer Control 2024; 31:10732748241299072. [PMID: 39487853 PMCID: PMC11531673 DOI: 10.1177/10732748241299072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/28/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Abnormalities in mitochondrial structure or function are closely related to the development of malignant tumors. Mitochondrial metabolic reprogramming provides precursor substances and energy for the vital activities of tumor cells, so that cancer cells can rapidly adapt to the unfavorable environment of hypoxia and nutrient deficiency. Mitochondria can enable tumor cells to gain the ability to proliferate, escape immune responses, and develop drug resistance by altering constitutive junctions, oxidative phosphorylation, oxidative stress, and mitochondrial subcellular relocalization. This greatly reduces the rate of effective clinical control of tumors. PURPOSE Explore the major role of mitochondria in cancer, as well as targeted mitochondrial therapies and mitochondria-associated markers. CONCLUSIONS This review provides a comprehensive analysis of the various aspects of mitochondrial aberrations and addresses drugs that target mitochondrial therapy, providing a basis for clinical mitochondria-targeted anti-tumor therapy.
Collapse
Affiliation(s)
- Zhiru Li
- Graduate School, North China University of Science and Technology, Tangshan, China
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Wu Zhang
- Center of Treatment of Myasthenia Gravis, People’s Hospital of Shijiazhuang Affiliated to Hebei Medical, Shijiazhuang, China
| | - Shaowei Guo
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Guoyan Qi
- Center of Treatment of Myasthenia Gravis, People’s Hospital of Shijiazhuang Affiliated to Hebei Medical, Shijiazhuang, China
| | - Jiandi Huang
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
- Graduate School, Hebei North University, Zhangjiakou, China
| | - Jin Gao
- Department of Thyroid & Breast Surgery Ward, Hebei General Hospital, Shijiazhuang, China
| | - Jing Zhao
- The Sixth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Lin Kang
- Department of Pathology, Hebei General Hospital, Shijiazhuang, China
| | - Qingxia Li
- The Fourth Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
35
|
Sanders Z, Moffitt BA, Treaster M, Larkins A, Khulordava N, Benjock J, Spencer J, Henrie K, Wurst MJ, Broom A, Tamez N, DeRosa G, Campbell M, Keller E, Powell A, Weinbrenner D, Abenavoli L, Edenfield WJ, Chung K, Boccuto L, Ivankovic D. Effects of Origanum majorana on Breast Cancer Cells: An Alternative to Chemotherapy? Metabolites 2023; 13:1083. [PMID: 37887408 PMCID: PMC10608860 DOI: 10.3390/metabo13101083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
Recent studies have reported several beneficial effects of natural compounds on cancerous cells, highlighting their use for future treatments. These preliminary findings have encouraged experiments with natural substances, such as plant extracts, to examine both cytotoxic and mitogenic effects and find alternative treatments for diseases such as breast cancer. This study examines the effects of microwave-assisted and ethanol maceration of marjoram (Origanum majorana) on MCF-7 breast cancer cell lines and normal breast tissue cell lines used as controls. Marjoram extracts displayed a cytotoxic effect on the MCF-7 cell lines and a mitogenic effect on the control cell lines at the MTS test. The metabolic profiles of MCF-7 and control cell lines were also assessed using the Biolog Phenotype Mammalian Metabolic (PM-M) platform and revealed statistically significant differences in the utilization of energy sources, metabolic activity in the presence of certain ionic species, and responses to metabolic effectors, such as stimulant/catabolic compounds and steroid hormones. Exposure to marjoram extracts exerted positive effects on the MCF-7 cells on the abnormal utilization of energy sources and the responses to metabolic effectors, while no major effects were detected on control cells. These effects were compared to the metabolic impact of the chemotherapeutic agent doxorubicin, which showed profound cytotoxic effects on both cancerous and normal breast cells. In conclusion, our in vitro evidence indicates that marjoram extracts are a promising alternative to chemotherapy in breast cancer since they can successfully eliminate cancerous cells by affecting their metabolic capacity to proliferate without inducing noticeable adverse effects on normal breast tissue.
Collapse
Affiliation(s)
- Zoe Sanders
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Bridgette A. Moffitt
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Madeleine Treaster
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Ashley Larkins
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Nicholas Khulordava
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Jennifer Benjock
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Jillian Spencer
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Krista Henrie
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Matthew J. Wurst
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Abigail Broom
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Noah Tamez
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Gianna DeRosa
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - McKenzie Campbell
- Department of Chemistry, Anderson University, Anderson, SC 29621, USA
| | - Elizabeth Keller
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Addison Powell
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
| | - Donna Weinbrenner
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa—Germaneto, 88100 Catanzaro, Italy;
| | | | - Ki Chung
- Prisma Health Cancer Institute, Greenville, SC 29605, USA (K.C.)
| | - Luigi Boccuto
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
| | - Diana Ivankovic
- Departments of Biology, Center for Cancer Research, Anderson University, 316 Boulevard, Anderson, SC 29621, USA; (Z.S.); (A.B.); (A.P.)
- Department of Healthcare Genetics, School of Nursing, Clemson University, Clemson, SC 29634, USA (J.S.); (L.B.)
- Department of Biological Sciences, Anderson University, 316 Boulevard, Anderson, SC 29621, USA
| |
Collapse
|