1
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Wang S, Wu L, Xie Y, Ge S, Wu Y, Chen L, Yi L, Yang J, Duan F, Huang L. Erjingpill bionic cerebrospinal fluid alleviates LPS-induced inflammatory response in BV2 cells by inhibiting glycolysis via mTOR. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118412. [PMID: 38824976 DOI: 10.1016/j.jep.2024.118412] [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: 04/13/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erjingpill, a well-known prescription documented in the classic Chinese medical text "Shengji Zonglu," has been proven to have effective alleviating effects on neuroinflammation in Alzheimer's disease (AD). Although the alterations in microglial cell glycolysis are known to play a crucial role in the development of neuroinflammation, it remains unclear whether the anti-neuroinflammatory effects of Erjingpill are associated with its impact on microglial cell glycolysis. AIM OF THE STUDY This study aims to determine whether Erjingpill exerts anti-neuroinflammatory effects by influencing microglial cell glycolysis. MATERIALS AND METHODS Firstly, Erjingpill decoction was prepared into an Erjingpill bionic cerebrospinal fluid (EBCF) through a process of in vitro intestinal absorption, hepatocyte incubation, and blood-brain barrier (BBB) transcytosis. Subsequently, UPLC/Q-TOF-MS/MS technology was used to analyze the compounds in Erjingpill and EBCF. Next, an in vitro neuroinflammation model was established by LPS-induced BV2 cells. The impact of EBCF on BV2 cell proliferation activity was evaluated using the CCK-8 assay, while the NO release was assessed using the Griess assay. Additionally, mRNA levels of pro-inflammatory factors (IL-1β, IL-6, TNF-α, and COX-2), anti-inflammatory factors (IL-10, IL-4, Arg-1, and TGF-β), M1 microglial markers (iNOS, CD86), M2 microglial markers (CD36, CD206), and glycolytic enzymes (HK2, GLUT1, PKM, and LDHA) were measured using qPCR. Furthermore, protein expression of microglial activation marker Iba-1, M1 marker iNOS, and M2 marker CD206 were identified through immunofluorescence, while concentrations of pro-inflammatory cytokines IL-1β and TNF-α were measured using ELISA. Enzymatic activity of glycolytic enzymes (HK, PK, and LDH) was assessed using assay kits, and the protein levels of pro-inflammatory factors (IL-1β, iNOS, and COX-2), anti-inflammatory factors (IL-10 and Arg-1), and key glycolytic proteins GLUT1 and PI3K/AKT/mTOR were detected by Western blot. RESULTS Through the analysis of Erjingpill and EBCF, 144 compounds were identified in Erjingpill and 40 compounds were identified in EBCF. The results demonstrated that EBCF effectively inhibited the elevation of inflammatory factors and glycolysis levels in LPS-induced BV2 cells, promoted polarization of M1 microglial cells towards the M2 phenotype, and suppressed the PI3K/AKT/mTOR inflammatory pathway. Moreover, EBCF alleviated LPS-induced BV2 cell inflammatory response by modulating mTOR to inhibit glycolysis. CONCLUSIONS EBCF exhibits significant anti-neuroinflammatory effects, likely attributed to its modulation of mTOR to inhibit microglial cell glycolysis. This study furnishes experimental evidence supporting the clinical utilization of Erjingpill for preventing and treating AD.
Collapse
Affiliation(s)
- Shuaikang Wang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Li Wu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Yongyan Xie
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Shuchao Ge
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Yi Wu
- Jiangxi Provincial Institute of Food and Drug Inspection and Testing, Nanchang, Jiangxi, 330004, China.
| | - Liping Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Longgen Yi
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Jie Yang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Feipeng Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Liping Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, China; Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| |
Collapse
|
3
|
Kitagawa Y, Kobayashi A, Cahill DP, Wakimoto H, Tanaka S. Molecular biology and novel therapeutics for IDH mutant gliomas: The new era of IDH inhibitors. Biochim Biophys Acta Rev Cancer 2024; 1879:189102. [PMID: 38653436 DOI: 10.1016/j.bbcan.2024.189102] [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: 12/14/2023] [Revised: 03/25/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
Gliomas with Isocitrate dehydrogenase (IDH) mutation represent a discrete category of primary brain tumors with distinct and unique characteristics, behaviors, and clinical disease outcomes. IDH mutations lead to aberrant high-level production of the oncometabolite D-2-hydroxyglutarate (D-2HG), which act as a competitive inhibitor of enzymes regulating epigenetics, signaling pathways, metabolism, and various other processes. This review summarizes the significance of IDH mutations, resulting upregulation of D-2HG and the associated molecular pathways in gliomagenesis. With the recent finding of clinically effective IDH inhibitors in these gliomas, this article offers a comprehensive overview of the new era of innovative therapeutic approaches based on mechanistic rationales, encompassing both completed and ongoing clinical trials targeting gliomas with IDH mutations.
Collapse
Affiliation(s)
- Yosuke Kitagawa
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA; Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA; Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, 1138655 Bunkyo-ku, Tokyo, Japan
| | - Ami Kobayashi
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA; Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA; Translational Neuro-Oncology Laboratory, Massachusetts General Hospital, Harvard Medical School, 02114 Boston, MA, USA.
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 7008558, Okayama, Japan
| |
Collapse
|
4
|
Jiang Z, He J, Zhang B, Wang L, Long C, Zhao B, Yang Y, Du L, Luo W, Hu J, Hong X. A Potential "Anti-Warburg Effect" in Circulating Tumor Cell-mediated Metastatic Progression? Aging Dis 2024; 16:AD.2023.1227. [PMID: 38300633 PMCID: PMC11745448 DOI: 10.14336/ad.2023.1227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Metabolic reprogramming is a defining hallmark of cancer metastasis, warranting thorough exploration. The tumor-promoting function of the "Warburg Effect", marked by escalated glycolysis and restrained mitochondrial activity, is widely acknowledged. Yet, the functional significance of mitochondria-mediated oxidative phosphorylation (OXPHOS) during metastasis remains controversial. Circulating tumor cells (CTCs) are considered metastatic precursors that detach from primary or secondary sites and harbor the potential to seed distant metastases through hematogenous dissemination. A comprehensive metabolic characterization of CTCs faces formidable obstacles, including the isolation of these rare cells from billions of blood cells, coupled with the complexities of ex vivo-culturing of CTC lines or the establishment of CTC-derived xenograft models (CDX). This review summarized the role of the "Warburg Effect" in both tumorigenesis and CTC-mediated metastasis. Intriguingly, bioinformatic analysis of single-CTC transcriptomic studies unveils a potential OXPHOS dominance over Glycolysis signature genes across several important cancer types. From these observations, we postulate a potential "Anti-Warburg Effect" (AWE) in CTCs-a metabolic shift bridging primary tumors and metastases. The observed AWE could be clinically important as they are significantly correlated with therapeutic response in melanoma and prostate patients. Thus, unraveling dynamic metabolic regulations within CTC populations might reveal an additional layer of regulatory complexities of cancer metastasis, providing an avenue for innovative anti-metastasis therapies.
Collapse
Affiliation(s)
- Zhuofeng Jiang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Jiapeng He
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Binyu Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Liping Wang
- Department of Oncology, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, China.
| | - Chunhao Long
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Boxi Zhao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Yufan Yang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Longxiang Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Weiren Luo
- Cancer Research Institute, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen Third People's Hospital, National Clinical Research Center for Infectious Diseases, Shenzhen, China.
| | - Jianyang Hu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Xin Hong
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Liu S, Benito‐Martin A, Pelissier Vatter FA, Hanif SZ, Liu C, Bhardwaj P, Sethupathy P, Farghli AR, Piloco P, Paik P, Mushannen M, Dong X, Otterburn DM, Cohen L, Bareja R, Krumsiek J, Cohen‐Gould L, Calto S, Spector JA, Elemento O, Lyden DC, Brown KA. Breast adipose tissue-derived extracellular vesicles from obese women alter tumor cell metabolism. EMBO Rep 2023; 24:e57339. [PMID: 37929643 PMCID: PMC10702795 DOI: 10.15252/embr.202357339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/07/2023] Open
Abstract
Breast adipose tissue is an important contributor to the obesity-breast cancer link. Extracellular vesicles (EVs) are nanosized particles containing selective cargo, such as miRNAs, that act locally or circulate to distant sites to modulate target cell functions. Here, we find that long-term education of breast cancer cells with EVs obtained from breast adipose tissue of women who are overweight or obese (O-EVs) results in increased proliferation. RNA-seq analysis of O-EV-educated cells demonstrates increased expression of genes involved in oxidative phosphorylation, such as ATP synthase and NADH: ubiquinone oxidoreductase. O-EVs increase respiratory complex protein expression, mitochondrial density, and mitochondrial respiration in tumor cells. The mitochondrial complex I inhibitor metformin reverses O-EV-induced cell proliferation. Several miRNAs-miR-155-5p, miR-10a-3p, and miR-30a-3p-which promote mitochondrial respiration and proliferation, are enriched in O-EVs relative to EVs from lean women. O-EV-induced proliferation and mitochondrial activity are associated with stimulation of the Akt/mTOR/P70S6K pathway, and are reversed upon silencing of P70S6K. This study reveals a new facet of the obesity-breast cancer link with human breast adipose tissue-derived EVs causing metabolic reprogramming of breast cancer cells.
Collapse
Affiliation(s)
- Shuchen Liu
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- Department of Breast SurgeryThe Second Hospital of Shandong UniversityJinanChina
| | - Alberto Benito‐Martin
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- Facultad de Medicina, Unidad de Investigación BiomédicaUniversidad Alfonso X el Sabio (UAX)MadridSpain
| | | | - Sarah Z Hanif
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Catherine Liu
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Priya Bhardwaj
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary MedicineCornell UniversityIthacaNYUSA
| | - Alaa R Farghli
- Department of Biomedical Sciences, College of Veterinary MedicineCornell UniversityIthacaNYUSA
| | - Phoebe Piloco
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Paul Paik
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Malik Mushannen
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- Weill Cornell Medicine – QatarDohaQatar
| | - Xue Dong
- Department of SurgeryWeill Cornell MedicineNew YorkNYUSA
| | | | - Leslie Cohen
- Department of SurgeryWeill Cornell MedicineNew YorkNYUSA
| | - Rohan Bareja
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNYUSA
| | - Jan Krumsiek
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNYUSA
| | - Leona Cohen‐Gould
- Department of BiochemistryWeill Cornell MedicineNew YorkNYUSA
- Core Laboratories CenterWeill Cornell MedicineNew YorkNYUSA
| | - Samuel Calto
- Department of Cognitive ScienceUniversity of California San DiegoLa JollaCAUSA
| | | | - Olivier Elemento
- Department of Physiology and BiophysicsWeill Cornell MedicineNew YorkNYUSA
| | - David C Lyden
- Departments of Pediatrics and Cell and Developmental BiologyWeill Cornell MedicineNew YorkNYUSA
| | - Kristy A Brown
- Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- Department of Cell Biology and PhysiologyUniversity of Kansas Medical CenterKansas CityKSUSA
- University of Kansas Cancer CenterKansas CityKSUSA
| |
Collapse
|
6
|
Xue KH, Jiang YF, Bai JY, Zhang DZ, Chen YH, Ma JB, Zhu ZJ, Wang X, Guo P. Melatonin suppresses Akt/mTOR/S6K activity, induces cell apoptosis, and synergistically inhibits cell growth with sunitinib in renal carcinoma cells via reversing Warburg effect. Redox Rep 2023; 28:2251234. [PMID: 37642220 PMCID: PMC10472857 DOI: 10.1080/13510002.2023.2251234] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Metabolic alteration drives renal cell carcinoma (RCC) development, while the impact of melatonin (MLT), a neurohormone secreted during darkness, on RCC cell growth and underlying mechanisms remains unclear. METHODS We detected concentration of metabolites through metabolomic analyses using UPLC-MS/MS, and the oxygen consumption rate was determined using the Seahorse Extracellular Flux analyzer. RESULTS We observed that MLT effectively inhibited RCC cell growth both in vitro and in vivo. Additionally, MLT increased ROS levels, suppressed antioxidant enzyme activity, and induced apoptosis. Furthermore, MLT treatment upregulated key TCA cycle metabolites while reducing aerobic glycolysis products, leading to higher oxygen consumption rate, ATP production, and membrane potential. Moreover, MLT treatment suppressed phosphorylation of Akt, mTOR, and p70 S6 Kinase as well as the expression of HIF-1α/VEGFA in RCC cells; these effects were reversed by NAC (ROS inhibitors). Conversely, MLT synergistically inhibited cell growth with sunitinib and counteracted the Warburg effect induced by sunitinib in RCC cells. CONCLUSIONS In conclusion, our results indicate that MLT treatment reverses the Warburg effect and promotes intracellular ROS production, which leads to the suppression of Akt/mTOR/S6K signaling pathway, induction of cell apoptosis, and synergistically inhibition of cell growth with sunitinib in RCC cells. Overall, this study provides new insights into the mechanisms underlying anti-tumor effect of MLT in RCC cells, and suggests that MLT might be a promising therapeutic for RCC.
Collapse
Affiliation(s)
- Kai-Hua Xue
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yi-Fan Jiang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Ji-Yu Bai
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Di-Ze Zhang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Yu-Hang Chen
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Jian-Bin Ma
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Zhi-Jing Zhu
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi’an, People’s Republic of China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, People’s Republic of China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi’an, People’s Republic of China
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an, People’s Republic of China
| |
Collapse
|
7
|
Liu M, Ji W, Zhao X, Liu X, Hu JF, Cui J. Therapeutic potential of engineering the mitochondrial genome. Biochim Biophys Acta Mol Basis Dis 2023:166804. [PMID: 37429560 DOI: 10.1016/j.bbadis.2023.166804] [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: 04/20/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
Mitochondrial diseases are a group of clinical disorders caused by mutations in the genes encoded by either the nuclear or the mitochondrial genome involved in mitochondrial oxidative phosphorylation. Disorders become evident when mitochondrial dysfunction reaches a cell-specific threshold. Similarly, the severity of disorders is related to the degree of gene mutation. Clinical treatments for mitochondrial diseases mainly rely on symptomatic management. Theoretically, replacing or repairing dysfunctional mitochondria to acquire and preserve normal physiological functions should be effective. Significant advances have been made in gene therapies, including mitochondrial replacement therapy, mitochondrial genome manipulation, nuclease programming, mitochondrial DNA editing, and mitochondrial RNA interference. In this paper, we review the recent progress in these technologies by focusing on advancements that overcome limitations.
Collapse
Affiliation(s)
- Mengmeng Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Wei Ji
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Xin Zhao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Xiaoliang Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Ji-Fan Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China; Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| | - Jiuwei Cui
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China.
| |
Collapse
|
8
|
Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15041192. [PMID: 36831534 PMCID: PMC9953833 DOI: 10.3390/cancers15041192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the "powerhouse" of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.
Collapse
|
9
|
Liu S, Benito-Martin A, Pelissier Vatter FA, Hanif SZ, Liu C, Bhardwaj P, Sethupathy P, Farghli AR, Piloco P, Paik P, Mushannen M, Otterburn DM, Cohen L, Bareja R, Krumsiek J, Cohen-Gould L, Calto S, Spector JA, Elemento O, Lyden D, Brown KA. Breast adipose tissue-derived extracellular vesicles from women with obesity stimulate mitochondrial-induced dysregulated tumor cell metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527715. [PMID: 36798307 PMCID: PMC9934680 DOI: 10.1101/2023.02.08.527715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Breast adipose tissue is an important contributor to the obesity-breast cancer link. Dysregulated cell metabolism is now an accepted hallmark of cancer. Extracellular vesicles (EVs) are nanosized particles containing selective cargo, such as miRNAs, that act locally or circulate to distant sites to modulate target cell functions. Here, we found that long-term education of breast cancer cells (MCF7, T47D) with EVs from breast adipose tissue of women who are overweight or obese (O-EVs) leads to sustained increased proliferative potential. RNA-Seq of O-EV-educated cells demonstrates increased expression of genes, such as ATP synthase and NADH: ubiquinone oxidoreductase, involved in oxidative phosphorylation. O-EVs increase respiratory complex protein expression, mitochondrial density, and mitochondrial respiration in tumor cells. Mitochondrial complex I inhibitor, metformin, reverses O-EV-induced cell proliferation. Several miRNAs, miR-155-5p, miR-10a-3p, and miR-30a-3p, which promote mitochondrial respiration and proliferation, are enriched in O-EVs relative to EVs from lean women. O-EV-induced proliferation and mitochondrial activity are associated with stimulation of the Akt/mTOR/P70S6K pathway, and are reversed upon silencing of P70S6K. This study reveals a new facet of the obesity-breast cancer link with human breast adipose tissue-derived EVs causing the metabolic reprogramming of ER+ breast cancer cells.
Collapse
|
10
|
Yang L, Xiong J, Li S, Liu X, Deng W, Liu W, Fu B. Mitochondrial metabolic reprogramming-mediated immunogenic cell death reveals immune and prognostic features of clear cell renal cell carcinoma. Front Oncol 2023; 13:1146657. [PMID: 37213288 PMCID: PMC10196130 DOI: 10.3389/fonc.2023.1146657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/11/2023] [Indexed: 05/23/2023] Open
Abstract
Background Mitochondrial metabolic reprogramming (MMR)-mediated immunogenic cell death (ICD) is closely related to the tumor microenvironment (TME). Our purpose was to reveal the TME characteristics of clear cell renal cell carcinoma (ccRCC) by using them. Methods Target genes were obtained by intersecting ccRCC differentially expressed genes (DEGs, tumor VS normal) with MMR and ICD-related genes. For the risk model, univariate COX regression and K-M survival analysis were used to identify genes most associated with overall survival (OS). Differences in the TME, function, tumor mutational load (TMB), and microsatellite instability (MSI) between high and low-risk groups were subsequently compared. Using risk scores and clinical variables, a nomogram was constructed. Predictive performance was evaluated by calibration plots and receiver operating characteristics (ROC). Results We screened 140 DEGs, including 12 prognostic genes for the construction of risk models. We found that the immune score, immune cell infiltration abundance, and TMB and MSI scores were higher in the high-risk group. Thus, high-risk populations would benefit more from immunotherapy. We also identified the three genes (CENPA, TIMP1, and MYCN) as potential therapeutic targets, of which MYCN is a novel biomarker. Additionally, the nomogram performed well in both TCGA (1-year AUC=0.862) and E-MTAB-1980 cohorts (1-year AUC=0.909). Conclusions Our model and nomogram allow accurate prediction of patients' prognoses and immunotherapy responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bin Fu
- *Correspondence: Bin Fu, ; Weipeng Liu,
| |
Collapse
|
11
|
Sollazzo M, De Luise M, Lemma S, Bressi L, Iorio M, Miglietta S, Milioni S, Kurelac I, Iommarini L, Gasparre G, Porcelli AM. Respiratory Complex I dysfunction in cancer: from a maze of cellular adaptive responses to potential therapeutic strategies. FEBS J 2022; 289:8003-8019. [PMID: 34606156 PMCID: PMC10078660 DOI: 10.1111/febs.16218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/03/2021] [Accepted: 10/01/2021] [Indexed: 01/14/2023]
Abstract
Mitochondria act as key organelles in cellular bioenergetics and biosynthetic processes producing signals that regulate different molecular networks for proliferation and cell death. This ability is also preserved in pathologic contexts such as tumorigenesis, during which bioenergetic changes and metabolic reprogramming confer flexibility favoring cancer cell survival in a hostile microenvironment. Although different studies epitomize mitochondrial dysfunction as a protumorigenic hit, genetic ablation or pharmacological inhibition of respiratory complex I causing a severe impairment is associated with a low-proliferative phenotype. In this scenario, it must be considered that despite the initial delay in growth, cancer cells may become able to resume proliferation exploiting molecular mechanisms to overcome growth arrest. Here, we highlight the current knowledge on molecular responses activated by complex I-defective cancer cells to bypass physiological control systems and to re-adapt their fitness during microenvironment changes. Such adaptive mechanisms could reveal possible novel molecular players in synthetic lethality with complex I impairment, thus providing new synergistic strategies for mitochondrial-based anticancer therapy.
Collapse
Affiliation(s)
- Manuela Sollazzo
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Monica De Luise
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Silvia Lemma
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Licia Bressi
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Iorio
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Stefano Miglietta
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Sara Milioni
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Ivana Kurelac
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Luisa Iommarini
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Giuseppe Gasparre
- Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Maria Porcelli
- Department of Pharmacy and Biotechnology (FABIT), Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy.,Centro di Studio e Ricerca sulle Neoplasie (CSR) Ginecologiche, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Interdepartmental Center for Industrial Research (CIRI) Life Sciences and Technologies for Health, Alma Mater Studiorum-University of Bologna, Ozzano dell'Emilia, Italy
| |
Collapse
|
12
|
Wang Q, Lu M, Zhu X, Gu X, Zhang T, Xia C, Yang L, Xu Y, Zhou M. The role of microglia immunometabolism in neurodegeneration: Focus on molecular determinants and metabolic intermediates of metabolic reprogramming. Biomed Pharmacother 2022; 153:113412. [DOI: 10.1016/j.biopha.2022.113412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
|
13
|
Xie L, Huang R, Huang H, Liu X, Yu J. Transcriptomics and Metabolomics Identify Drug Resistance of Dormant Cell in Colorectal Cancer. Front Pharmacol 2022; 13:879751. [PMID: 35462906 PMCID: PMC9024109 DOI: 10.3389/fphar.2022.879751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Tumor dormancy is an important way to develop drug resistance. This study aimed to identify the characteristics of colorectal cancer (CRC) cell dormancy. Methods: Based on the CRC cohorts, a total of 1,044 CRC patients were included in this study, and divided into a dormant subgroup and proliferous subgroup. Non-negative matrix factorization (NMF) was used to distinguish the dormant subgroup of CRC via transcriptome data of cancer tissues. Gene Set Enrichment Analysis (GSEA) was used to explore the characteristics of dormant CRC. The characteristics were verified in the cell model, which was used to predict key factors driving CRC dormancy. Potential treatments for CRC dormancy were also examined. Results: The dormant subgroup had a poor prognosis and was more likely to relapse. GSEA analysis showed two defining characteristics of the dormant subgroup, a difference in energy metabolism and synergistic effects of cancer-associated fibroblasts (CAFs), which were verified in a dormant cell model. Transcriptome and clinical data identified LMOD1, MAB21L2, and ASPN as important factors associated with cell dormancy and verified that erlotinib, and CB-839 were potential treatment options. Conclusion: Dormant CRC is associated with high glutamine metabolism and synergizes with CAFs in 5-FU resistance, and the key effectors are LMOD1, MAB21L2, and ASPN. Austocystin D, erlotinib, and CB-839 may be useful for dormant CRC.
Collapse
Affiliation(s)
- Lang Xie
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Renli Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongyun Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital (Guangdong Gastrointestinal and Anal Hospital), Sun Yat-sen University, Guangzhou, China
- *Correspondence: Xiaoxia Liu, ; Jinlong Yu,
| | - Jinlong Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Xiaoxia Liu, ; Jinlong Yu,
| |
Collapse
|
14
|
Herst PM, Carson GM, Eccles DA, Berridge MV. Bioenergetic and Metabolic Adaptation in Tumor Progression and Metastasis. Front Oncol 2022; 12:857686. [PMID: 35372069 PMCID: PMC8968714 DOI: 10.3389/fonc.2022.857686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
The ability of cancer cells to adjust their metabolism in response to environmental changes is a well-recognized hallmark of cancer. Diverse cancer and non-cancer cells within tumors compete for metabolic resources. Metabolic demands change frequently during tumor initiation, progression and metastasis, challenging our quest to better understand tumor biology and develop novel therapeutics. Vascularization, physical constraints, immune responses and genetic instability promote tumor evolution resulting in immune evasion, opportunities to breach basement membrane barriers and spread through the circulation and lymphatics. In addition, the unfolded protein response linked to the ubiquitin proteasome system is a key player in addressing stoichiometric imbalances between nuclear and mitochondrially-encoded protein subunits of respiratory complexes, and nuclear-encoded mitochondrial ribosomal protein subunits. While progressive genetic changes, some of which affect metabolic adaptability, contribute to tumorigenesis and metastasis through clonal expansion, epigenetic changes are also important and more dynamic in nature. Understanding the role of stromal and immune cells in the tumor microenvironment in remodeling cancer cell energy metabolism has become an increasingly important area of research. In this perspective, we discuss the adaptations made by cancer cells to balance mitochondrial and glycolytic energy metabolism. We discuss how hypoxia and nutrient limitations affect reductive and oxidative stress through changes in mitochondrial electron transport activity. We propose that integrated responses to cellular stress in cancer cells are central to metabolic flexibility in general and bioenergetic adaptability in particular and are paramount in tumor progression and metastasis.
Collapse
Affiliation(s)
- Patries M. Herst
- Department of Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
- Department of Radiation Therapy, University of Otago, Wellington, New Zealand
| | - Georgia M. Carson
- Department of Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - David A. Eccles
- Department of Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Michael V. Berridge
- Department of Cell Biology, Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
15
|
Lai HT, Canoy RJ, Campanella M, Vassetzky Y, Brenner C. Ca2+ Transportome and the Interorganelle Communication in Hepatocellular Carcinoma. Cells 2022; 11:cells11050815. [PMID: 35269437 PMCID: PMC8909868 DOI: 10.3390/cells11050815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of liver cancer with a poor prognosis for survival given the complications it bears on the patient. Though damages to the liver are acknowledged prodromic factors, the precise molecular aetiology remains ill-defined. However, many genes coding for proteins involved in calcium (Ca2+) homeostasis emerge as either mutated or deregulated. Ca2+ is a versatile signalling messenger that regulates functions that prime and drive oncogenesis, favouring metabolic reprogramming and gene expression. Ca2+ is present in cell compartments, between which it is trafficked through a network of transporters and exchangers, known as the Ca2+ transportome. The latter regulates and controls Ca2+ dynamics and tonicity. In HCC, the deregulation of the Ca2+ transportome contributes to tumorigenesis, the formation of metastasizing cells, and evasion of cell death. In this review, we reflect on these aspects by summarizing the current knowledge of the Ca2+ transportome and overviewing its composition in the plasma membrane, endoplasmic reticulum, and the mitochondria.
Collapse
Affiliation(s)
- Hong-Toan Lai
- CNRS, Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (H.-T.L.); (R.J.C.); (M.C.); (Y.V.)
| | - Reynand Jay Canoy
- CNRS, Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (H.-T.L.); (R.J.C.); (M.C.); (Y.V.)
- Institute of Human Genetics, National Institutes of Health, University of the Philippines, Manila 1000, Philippines
| | - Michelangelo Campanella
- CNRS, Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (H.-T.L.); (R.J.C.); (M.C.); (Y.V.)
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London NW1 0TU, UK
- Consortium for Mitochondrial Research, University College London, London WC1 0TU, UK
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Yegor Vassetzky
- CNRS, Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (H.-T.L.); (R.J.C.); (M.C.); (Y.V.)
| | - Catherine Brenner
- CNRS, Institut Gustave Roussy, Aspects Métaboliques et Systémiques de l’Oncogénèse pour de Nouvelles Approches Thérapeutiques, Université Paris-Saclay, 94805 Villejuif, France; (H.-T.L.); (R.J.C.); (M.C.); (Y.V.)
- Correspondence:
| |
Collapse
|
16
|
Bao L, Xu T, Lu X, Huang P, Pan Z, Ge M. Metabolic Reprogramming of Thyroid Cancer Cells and Crosstalk in Their Microenvironment. Front Oncol 2021; 11:773028. [PMID: 34926283 PMCID: PMC8674491 DOI: 10.3389/fonc.2021.773028] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/05/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolism differs significantly between tumor and normal cells. Metabolic reprogramming in cancer cells and metabolic interplay in the tumor microenvironment (TME) are important for tumor formation and progression. Tumor cells show changes in both catabolism and anabolism. Altered aerobic glycolysis, known as the Warburg effect, is a well-recognized characteristic of tumor cell energy metabolism. Compared with normal cells, tumor cells consume more glucose and glutamine. The enhanced anabolism in tumor cells includes de novo lipid synthesis as well as protein and nucleic acid synthesis. Although these forms of energy supply are uneconomical, they are required for the functioning of cancer cells, including those in thyroid cancer (TC). Increasing attention has recently focused on alterations of the TME. Understanding the metabolic changes governing the intricate relationship between TC cells and the TME may provide novel ideas for the treatment of TC.
Collapse
Affiliation(s)
- Lisha Bao
- Second Clinical College, Zhejiang Chinese Medical School, Hangzhou, China
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Tong Xu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xixuan Lu
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ping Huang
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zongfu Pan
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Minghua Ge
- ENT-Head & Neck Surgery Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
17
|
Gao M, Xie Y, Lei K, Zhao Y, Kurum A, Van Herck S, Guo Y, Hu X, Tang L. A Manganese Phosphate Nanocluster Activates the cGAS‐STING Pathway for Enhanced Cancer Immunotherapy. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100065] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Min Gao
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yu‐Qing Xie
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Kewen Lei
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yu Zhao
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Armand Kurum
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Simon Van Herck
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Yugang Guo
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Xiaomeng Hu
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| | - Li Tang
- Institute of Bioengineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
- Institute of Materials Science and Engineering École Polytechnique Fédérale de Lausanne (EPFL) Lausanne 1015 Switzerland
| |
Collapse
|
18
|
Yang S, Qin C, Hu ZW, Zhou LQ, Yu HH, Chen M, Bosco DB, Wang W, Wu LJ, Tian DS. Microglia reprogram metabolic profiles for phenotype and function changes in central nervous system. Neurobiol Dis 2021; 152:105290. [PMID: 33556540 DOI: 10.1016/j.nbd.2021.105290] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 12/31/2020] [Accepted: 02/03/2021] [Indexed: 12/13/2022] Open
Abstract
In response to various types of environmental and cellular stress, microglia rapidly activate and exhibit either pro- or anti-inflammatory phenotypes to maintain tissue homeostasis. Activation of microglia can result in changes in morphology, phagocytosis capacity, and secretion of cytokines. Furthermore, microglial activation also induces changes to cellular energy demand, which is dependent on the metabolism of various metabolic substrates including glucose, fatty acids, and amino acids. Accumulating evidence demonstrates metabolic reprogramming acts as a key driver of microglial immune response. For instance, microglia in pro-inflammatory states preferentially use glycolysis for energy production, whereas, cells in anti-inflammatory states are mainly powered by oxidative phosphorylation and fatty acid oxidation. In this review, we summarize recent findings regarding microglial metabolic pathways under physiological and pathological circumtances. We will then discuss how metabolic reprogramming can orchestrate microglial response to a variety of central nervous system pathologies. Finally, we highlight how manipulating metabolic pathways can reprogram microglia towards beneficial functions, and illustrate the therapeutic potential for inflammation-related neurological diseases.
Collapse
Affiliation(s)
- Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zi-Wei Hu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hai-Han Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, United States of America
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, United States of America.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Szabo C. Hydrogen Sulfide, an Endogenous Stimulator of Mitochondrial Function in Cancer Cells. Cells 2021; 10:cells10020220. [PMID: 33499368 PMCID: PMC7911547 DOI: 10.3390/cells10020220] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) has a long history as toxic gas and environmental hazard; inhibition of cytochrome c oxidase (mitochondrial Complex IV) is viewed as a primary mode of its cytotoxic action. However, studies conducted over the last two decades unveiled multiple biological regulatory roles of H2S as an endogenously produced mammalian gaseous transmitter. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently viewed as the principal mammalian H2S-generating enzymes. In contrast to its inhibitory (toxicological) mitochondrial effects, at lower (physiological) concentrations, H2S serves as a stimulator of electron transport in mammalian mitochondria, by acting as an electron donor—with sulfide:quinone oxidoreductase (SQR) being the immediate electron acceptor. The mitochondrial roles of H2S are significant in various cancer cells, many of which exhibit high expression and partial mitochondrial localization of various H2S producing enzymes. In addition to the stimulation of mitochondrial ATP production, the roles of endogenous H2S in cancer cells include the maintenance of mitochondrial organization (protection against mitochondrial fission) and the maintenance of mitochondrial DNA repair (via the stimulation of the assembly of mitochondrial DNA repair complexes). The current article overviews the state-of-the-art knowledge regarding the mitochondrial functions of endogenously produced H2S in cancer cells.
Collapse
Affiliation(s)
- Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
20
|
Abstract
Platelet mitochondria can be used in the study of mitochondrial dysfunction in various complex diseases and can help in finding biological markers for diagnosing the disease, monitoring its course and the effects of treatment. The aim of this chapter was to describe in detail the method of measuring mitochondrial respiration in platelets using high-resolution respirometry. The described method was successfully used for the study of mitochondrial dysfunction in neuropsychiatric diseases.
Collapse
|
21
|
Zhao S, Zhang X, Shi Y, Cheng L, Song T, Wu B, Li J, Yang H. MIEF2 over-expression promotes tumor growth and metastasis through reprogramming of glucose metabolism in ovarian cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:286. [PMID: 33317572 PMCID: PMC7737286 DOI: 10.1186/s13046-020-01802-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/04/2020] [Indexed: 01/20/2023]
Abstract
Background Increasing evidence has revealed the close link between mitochondrial dynamic dysfunction and cancer. MIEF2 (mitochondrial elongation factor 2) is mitochondrial outer membrane protein that functions in the regulation of mitochondrial fission. However, the expression, clinical significance and biological functions of MIEF2 are still largely unclear in human cancers, especially in ovarian cancer (OC). Methods The expression and clinical significance of MIEF2 were determined by qRT-PCR, western blot and immunohistochemistry analyses in tissues and cell lines of OC. The biological functions of MIEF2 in OC were determined by in vitro and in vivo cell growth and metastasis assays. Furthermore, the effect of MIEF2 on metabolic reprogramming of OC was determined by metabolomics and glucose metabolism analyses. Results MIEF2 expression was significantly increased in OC mainly due to the down-regulation of miR-424-5p, which predicts poor survival for patients with OC. Knockdown of MIEF2 significantly suppressed OC cell growth and metastasis both in vitro and in vivo by inhibiting G1-S cell transition, epithelial-to-mesenchymal transition (EMT) and inducing cell apoptosis, while forced expression of MIEF2 had the opposite effects. Mechanistically, mitochondrial fragmentation-suppressed cristae formation and thus glucose metabolism switch from oxidative phosphorylation to glycolysis was found to be involved in the promotion of growth and metastasis by MIEF2 in OC cells. Conclusions MIEF2 plays a critical role in the progression of OC and may serve as a valuable prognostic biomarker and therapeutic target in the treatment of this malignancy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-020-01802-9.
Collapse
Affiliation(s)
- Shuhua Zhao
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China
| | - Xiaohong Zhang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China
| | - Yuan Shi
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China
| | - Lu Cheng
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China
| | - Tingting Song
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China
| | - Bing Wu
- Department of Geriatrics, the 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Jia Li
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China.
| | - Hong Yang
- Department of Gynaecology and Obstetrics, Xijing Hospital, Fourth Military Medical University, 15 Changle Western Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
22
|
A simple indirect colorimetric assay for measuring mitochondrial energy metabolism based on uncoupling sensitivity. Biochem Biophys Rep 2020; 24:100858. [PMID: 33294636 PMCID: PMC7691152 DOI: 10.1016/j.bbrep.2020.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/19/2020] [Accepted: 11/09/2020] [Indexed: 11/22/2022] Open
Abstract
Purpose Cancer cells rapidly adjust their balance between glycolytic and mitochondrial ATP production in response to changes in their microenvironment and to treatments like radiation and chemotherapy. Reliable, simple, high throughput assays that measure the levels of mitochondrial energy metabolism in cells are useful determinants of treatment effects. Mitochondrial metabolism is routinely determined by measuring the rate of oxygen consumption (OCR). We have previously shown that indirect inhibition of plasma membrane electron transport (PMET) by the mitochondrial uncoupler, FCCP, may also be a reliable measure of mitochondrial energy metabolism. Here, we aimed to validate these earlier findings by exploring the relationship between stimulation of oxygen consumption by FCCP and inhibition of PMET. Methods We measured PMET by reduction of the cell impermeable tetrazolium salt WST-1/PMS. We characterised the effect of different growth conditions on the extent of PMET inhibition by FCCP. Next, we compared FCCP-mediated PMET inhibition with FCCP-mediated stimulation of OCR using the Seahorse XF96e flux analyser, in a panel of cancer cell lines. Results We found a strong inverse correlation between stimulation of OCR and PMET inhibition by FCCP. PMET and OCR were much more severely affected by FCCP in cells that rely on mitochondrial energy production than in cells with a more glycolytic phenotype. Conclusion Indirect inhibition of PMET by FCCP is a reliable, simple and inexpensive high throughput assay to determine the level of mitochondrial energy metabolism in cancer cells. WST-1/PMS dye reduction measures NADH-driven plasma membrane electron transport. FCCP stimulates mitochondrial oxygen consumption and inhibits dye reduction. The FCCP effect on dye reduction and oxygen consumption is inversely correlated. FCCP-mediated inhibition of dye reduction is a measure of mitochondrial metabolism.
Collapse
|
23
|
Grasso C, Eccles DA, Boukalova S, Fabre MS, Dawson RH, Neuzil J, Herst PM, Berridge MV. Mitochondrial DNA Affects the Expression of Nuclear Genes Involved in Immune and Stress Responses in a Breast Cancer Model. Front Physiol 2020; 11:543962. [PMID: 33329014 PMCID: PMC7732479 DOI: 10.3389/fphys.2020.543962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 10/09/2020] [Indexed: 12/29/2022] Open
Abstract
Tumor cells without mitochondrial (mt) DNA (ρ0 cells) are auxotrophic for uridine, and their growth is supported by pyruvate. While ATP synthesis in ρ0 cells relies on glycolysis, they fail to form tumors unless they acquire mitochondria from stromal cells. Mitochondrial acquisition restores respiration that is essential for de novo pyrimidine biosynthesis and for mitochondrial ATP production. The physiological processes that underpin intercellular mitochondrial transfer to tumor cells lacking mtDNA and the metabolic remodeling and restored tumorigenic properties of cells that acquire mitochondria are not well understood. Here, we investigated the changes in mitochondrial and nuclear gene expression that accompany mtDNA deletion and acquisition in metastatic murine 4T1 breast cancer cells. Loss of mitochondrial gene expression in 4T1ρ0 cells was restored in cells recovered from subcutaneous tumors that grew from 4T1ρ0 cells following acquisition of mtDNA from host cells. In contrast, the expression of most nuclear genes that encode respiratory complex subunits and mitochondrial ribosomal subunits was not greatly affected by loss of mtDNA, indicating ineffective mitochondria-to-nucleus communication systems for these nuclear genes. Further, analysis of nuclear genes whose expression was compromised in 4T1ρ0 cells showed that immune- and stress-related genes were the most highly differentially expressed, representing over 70% of those with greater than 16-fold higher expression in 4T1 compared with 4T1ρ0 cells. The monocyte recruiting chemokine, Ccl2, and Psmb8, a subunit of the immunoproteasome that generates MHCI-binding peptides, were the most highly differentially expressed. Early monocyte/macrophage recruitment into the tumor mass was compromised in 4T1ρ0 cells but recovered before mtDNA could be detected. Taken together, our results show that mitochondrial acquisition by tumor cells without mtDNA results in bioenergetic remodeling and re-expression of genes involved in immune function and stress adaptation.
Collapse
Affiliation(s)
- Carole Grasso
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - David A. Eccles
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czechia
| | | | | | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czechia
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Patries M. Herst
- Malaghan Institute of Medical Research, Wellington, New Zealand
- Department of Radiation Therapy, University of Otago, Wellington, New Zealand
| | | |
Collapse
|
24
|
Medina MÁ. Metabolic Reprogramming is a Hallmark of Metabolism Itself. Bioessays 2020; 42:e2000058. [PMID: 32939776 DOI: 10.1002/bies.202000058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/13/2020] [Indexed: 12/16/2022]
Abstract
The reprogramming of metabolism has been identified as one of the hallmarks of cancer. It is becoming more and more frequent to connect other diseases with metabolic reprogramming. This article aims to argue that metabolic reprogramming is not driven by disease but instead is the main hallmark of metabolism, based on its dynamic behavior that allows it to continuously adapt to changes in the internal and external conditions.
Collapse
Affiliation(s)
- Miguel Ángel Medina
- Andalucía Tech, Facultad de Ciencias, Departamento de Biología Molecular y Bioquímica, and IBIMA (Biomedical Research Institute of Málaga), Universidad de Málaga, Málaga, E-29071, Spain.,CIBER de Enfermedades Raras (CIBERER), Málaga, E-29071, Spain
| |
Collapse
|
25
|
Muresanu C, Somasundaram SG, Vissarionov SV, Torres Solis LF, Solís Herrera A, Kirkland CE, Aliev G. Updated Understanding of Cancer as a Metabolic and Telomere-Driven Disease, and Proposal for Complex Personalized Treatment, a Hypothesis. Int J Mol Sci 2020; 21:E6521. [PMID: 32906638 PMCID: PMC7555410 DOI: 10.3390/ijms21186521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022] Open
Abstract
In this review, we propose a holistic approach to understanding cancer as a metabolic disease. Our search for relevant studies in medical databases concludes that cancer cells do not evolve directly from normal healthy cells. We hypothesize that aberrant DNA damage accumulates over time-avoiding the natural DNA controls that otherwise repair or replace the rapidly replicating cells. DNA damage starts to accumulate in non-replicating cells, leading to senescence and aging. DNA damage is linked with genetic and epigenetic factors, but the development of cancer is favored by telomerase activity. Evidence indicates that telomere length is affected by chronic inflammations, alterations of mitochondrial DNA, and various environmental factors. Emotional stress also influences telomere length. Chronic inflammation can cause oxidative DNA damage. Oxidative stress, in turn, can trigger mitochondrial changes, which ultimately alter nuclear gene expression. This vicious cycle has led several scientists to view cancer as a metabolic disease. We have proposed complex personalized treatments that seek to correct multiple changes simultaneously using a psychological approach to reduce chronic stress, immune checkpoint therapy with reduced doses of chemo and radiotherapy, minimal surgical intervention, if any, and mitochondrial metabolic reprogramming protocols supplemented by intermittent fasting and personalized dietary plans without interfering with the other therapies.
Collapse
Affiliation(s)
- Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478 Cluj-Napoca, Romania;
| | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Sergey V. Vissarionov
- The Department of Spinal Pathology and Neurosurgery, Turner Scientific and Research Institute for Children’s Orthopedics, Street Parkovskya 64-68, Pushkin, 196603 Saint-Petersburg, Russia;
| | | | | | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia
- Research Institute of Human Morphology, Russian Academy of Medical Science, Street Tsyurupa 3, 117418 Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432 Moscow, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
| |
Collapse
|
26
|
Zhao H, Yan G, Zheng L, Zhou Y, Sheng H, Wu L, Zhang Q, Lei J, Zhang J, Xin R, Jiang L, Zhang X, Chen Y, Wang J, Xu Y, Li D, Li Y. STIM1 is a metabolic checkpoint regulating the invasion and metastasis of hepatocellular carcinoma. Theranostics 2020; 10:6483-6499. [PMID: 32483465 PMCID: PMC7255033 DOI: 10.7150/thno.44025] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Cancer cells undergoing invasion and metastasis possess a phenotype with attenuated glycolysis, but enhanced fatty acid oxidation (FAO). Calcium (Ca2+)-mediated signaling pathways are implicated in tumor metastasis and metabolism regulation. Stromal-interaction molecule 1 (STIM1) triggered store-operated Ca2+ entry (SOCE) is the major route of Ca2+ influx for non-excitable cells including hepatocellular carcinoma (HCC) cells. However, whether and how STIM1 regulates the invasion and metastasis of HCC via metabolic reprogramming is unclear. Methods: The expressions of STIM1 and Snail1 in the HCC tissues and cells were measured by immunohistochemistry, Western-blotting and quantitative PCR. STIM1 knockout-HCC cells were generated by CRISPR-Cas9, and gene-overexpression was mediated via lentivirus transfection. Besides, the invasive and metastatic activities of HCC cells were assessed by transwell assay, anoikis rate in vitro and lung metastasis in vivo. Seahorse energy analysis and micro-array were used to evaluate the glucose and lipid metabolism. Results: STIM1 was down-regulated in metastatic HCC cells rather than in proliferating HCC cells, and low STIM1 levels were associated with poor outcome of HCC patients. During tumor growth, STIM1 stabilized Snail1 protein by activating the CaMKII/AKT/GSK-3β pathway. Subsequently, the upregulated Snail1 suppressed STIM1/SOCE during metastasis. STIM1 restoration significantly diminished anoikis-resistance and metastasis induced by Snail1. Mechanistically, the downregulated STIM1 shifted the anabolic/catabolic balance, i.e., from aerobic glycolysis towards AMPK-activated fatty acid oxidation (FAO), which contributed to Snail1-driven metastasis and anoikis-resistance. Conclusions: Our data provide the molecular basis that STIM1 orchestrates invasion and metastasis via reprogramming HCC metabolism.
Collapse
|
27
|
Ma DY, Lai Q, Peng KJ, Wang LL, Li ZX, Liu LJ, Luo ZY, Liu SY. Synthesis and anti-OXPHOS, antitumor activities of DLC modified spinosyn derivatives. Bioorg Med Chem Lett 2020; 30:127047. [PMID: 32139325 DOI: 10.1016/j.bmcl.2020.127047] [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: 01/15/2020] [Revised: 01/31/2020] [Accepted: 02/15/2020] [Indexed: 10/25/2022]
Abstract
A series of DLC (delocalized lipophilic cation) modified spinosyn derivatives were synthesized and evaluated for antitumor efficacies both in vitro and in vivo. Cancer cell based antiproliferative assays indicated that the more lipophilic derivatives had stronger inhibitory effects on the tested cancer cell lines. Compound 7b and 8b exhibited strong anti-OXPHOS and apoptosis inducing ability. Notable antitumor efficacies of 7b (5 mg/kg) and 8b (2.5 mg/kg) were observed in the in vivo tumor xenograft experiments, however, lethal toxicities were observed on higher dosages. Our findings indicated that DLC modification is a viable strategy to enhance the anti-OXPHOS and antitumor efficacies of spinosyn derivatives.
Collapse
Affiliation(s)
- Da-You Ma
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| | - Qin Lai
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Kun-Jian Peng
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Long-Long Wang
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Zeng-Xia Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Li-Jun Liu
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Zhi-Yong Luo
- School of Life Sciences, Central South University, Changsha 410013, China
| | - Su-You Liu
- School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
28
|
Commander R, Wei C, Sharma A, Mouw JK, Burton LJ, Summerbell E, Mahboubi D, Peterson RJ, Konen J, Zhou W, Du Y, Fu H, Shanmugam M, Marcus AI. Subpopulation targeting of pyruvate dehydrogenase and GLUT1 decouples metabolic heterogeneity during collective cancer cell invasion. Nat Commun 2020; 11:1533. [PMID: 32210228 PMCID: PMC7093428 DOI: 10.1038/s41467-020-15219-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Phenotypic heterogeneity exists within collectively invading packs of tumor cells, suggesting that cellular subtypes cooperate to drive invasion and metastasis. Here, we take a chemical biology approach to probe cell:cell cooperation within the collective invasion pack. These data reveal metabolic heterogeneity within invasive chains, in which leader cells preferentially utilize mitochondrial respiration and trailing follower cells rely on elevated glucose uptake. We define a pyruvate dehydrogenase (PDH) dependency in leader cells that can be therapeutically exploited with the mitochondria-targeting compound alexidine dihydrochloride. In contrast, follower cells highly express glucose transporter 1 (GLUT1), which sustains an elevated level of glucose uptake required to maintain proliferation. Co-targeting of both leader and follower cells with PDH and GLUT1 inhibitors, respectively, inhibits cell growth and collective invasion. Taken together, our work reveals metabolic heterogeneity within the lung cancer collective invasion pack and provides rationale for co-targeting PDH and GLUT1 to inhibit collective invasion. The presence of phenotypic heterogeneity in collectively invading cells suggests cooperation amongst distinct subtypes of cells to promote invasion and metastasis. Here, the authors use chemical biology tools and report metabolic heterogeneity within the lung cancer collective invasion pack.
Collapse
Affiliation(s)
- R Commander
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - C Wei
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - A Sharma
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - J K Mouw
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - L J Burton
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - E Summerbell
- Graduate Program in Cancer Biology, Emory University, Atlanta, GA, USA
| | - D Mahboubi
- Graduate Program in Molecular Systems Pharmacology, Emory University, Atlanta, GA, USA
| | - R J Peterson
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA, USA
| | - J Konen
- Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - W Zhou
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | - Y Du
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - H Fu
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA.,Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA, USA
| | - M Shanmugam
- Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| | - A I Marcus
- Winship Cancer Institute, Emory University, Atlanta, GA, USA. .,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
29
|
Tan Y, Shao R, Li J, Huang H, Wang Y, Zhang M, Cao J, Zhang J, Bu J. PITPNC1 fuels radioresistance of rectal cancer by inhibiting reactive oxygen species production. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:126. [PMID: 32175419 PMCID: PMC7049036 DOI: 10.21037/atm.2020.02.37] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 02/01/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neoadjuvant radiotherapy is a commonly used method for the current standard-of-care for most patients with rectal cancer, when the effects of radioresistance are limited. The phosphatidylinositol transfer protein, cytoplasmic 1 (PITPNC1), a lipid-metabolism-related gene, has previously been proved to manifest pro-cancer effects in multiple types of cancer. However, whether PITPNC1 plays a role for developing radioresistance in rectal cancer patients is still unknown. Therefore, this study aims to investigate the role of PITPNC1 in rectal cancer radioresistance. METHODS Patient-derived tissue were used to detect the difference in the expression level of PITPNC1 between radioresistant and radiosensitive patients. Bioinformatic analyses of high-throughput gene expression data were applied to uncover the correlations between PITPNC1 level and oxidative stress. Two rectal cancer cell lines, SW620, and HCT116, were selected in vitro to investigate the effect of PITPNC1 on radioresistance, reactive oxygen species (ROS) generation, apoptosis, and proliferation in rectal cancer. RESULTS PITPNC1 is highly expressed in radioresistant patient-derived rectal cancer tissues compared to radiosensitive tissue; therefore, PITPNC1 inhibits the generation of ROS and improves the extent of radioresistance of rectal cancer cell lines and then inhibits apoptosis. Knocking down PITPNC1 facilitates the production of ROS while application of the ROS scavenger, N-acetyl-L-cysteine (NAC), could reverse this effect. CONCLUSIONS PITPNC1 fuels radioresistance of rectal cancer via the inhibition of ROS generation.
Collapse
Affiliation(s)
- Yujing Tan
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ruoyang Shao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingyu Li
- Department of Pathology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Hongyun Huang
- Department of Abdominal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanru Wang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Menglan Zhang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianyun Cao
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junde Zhang
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junguo Bu
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
30
|
Abstract
Apart from reliable management of the "powerhouse" of the cell, mitochondria faithfully orchestrate a diverse array of important and critical functions in governing cellular signaling, apoptosis, autophagy, mitophagy and innate and adaptive immune system. Introduction of instability and imbalance in the mitochondrial own genome or the nuclear encoded mitochondrial proteome would result in the manifestation of various diseases through alterations in the oxidative phosphorylation system (OXPHOS) and nuclear-mitochondria retrograde signaling. Understanding mitochondrial biology and dynamism are thus of paramount importance to develop strategies to prevent or treat various diseases caused due to mitochondrial alterations.
Collapse
Affiliation(s)
- Santanu Dasgupta
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| |
Collapse
|
31
|
Discovery of a Ruthenium Complex for the Theranosis of Glioma through Targeting the Mitochondrial DNA with Bioinformatic Methods. Int J Mol Sci 2019; 20:ijms20184643. [PMID: 31546801 PMCID: PMC6770666 DOI: 10.3390/ijms20184643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Glioma is the most aggressive and lethal brain tumor in humans. Mutations of mitochondrial DNA (mtDNA) are commonly found in tumor cells and are closely associated with tumorigenesis and progress. However, glioma-specific inhibitors that reflect the unique feature of tumor cells are rare. Here we uncover RC-7, a ruthenium complex with strong red fluorescence, could bind with glioma mtDNA and then inhibited the growth of human glioma cells but not that of neuronal cells, liver, or endothelial cells. RC-7 significantly reduced energy production and increased the oxidative stress in the glioma cells. Administration of RC-7 into mice not only could be observed in the glioma mass of brain by fluorescence imaging, but also obviously prevented the growth of xenograft glioma and prolonged mouse survival days. The findings suggested the theranostic application of a novel type of complex through targeting the tumor mtDNA.
Collapse
|
32
|
PLCε regulates prostate cancer mitochondrial oxidative metabolism and migration via upregulation of Twist1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:337. [PMID: 31383001 PMCID: PMC6683382 DOI: 10.1186/s13046-019-1323-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/14/2019] [Indexed: 12/17/2022]
Abstract
Background Metabolic rewiring is a common feature of many cancer types, including prostate cancer (PCa). Alterations in master genes lead to mitochondrial metabolic rewiring and provide an appealing target to inhibit cancer progression and improve survival. Phospholipase C (PLC)ε is a regulator of tumor generation and progression. However, its role in mitochondrial metabolism remains unclear. Methods The GEO, The Cancer Genome Atlas, and the GTEx databases were used to determine Twist1 mRNA levels in tumors and their non-tumor counterparts. Fifty-five PCa and 48 benign prostatic hypertrophy tissue samples were tested for the presence of PLCε and Twist1 immunohistochemically. An association between PLCε and Twist1 was determined by Pearson’s correlation analysis. PLCε was knocked down with a lentiviral short hairpin RNA. Mitochondrial activity was assessed by measuring the oxygen consumption rate. Western blotting analyses were used to measure levels of PPARβ, Twist1, phosphorylated (p)-Twist1, p-MEK, p-ERK, p-P38, and p-c-Jun N-terminal kinase (JNK). Cells were treated with inhibitors of MEK, JNK, and P38 MAPK, and an agonist and inhibitor of peroxisome proliferator activated receptor (PPAR) β, to evaluate which signaling pathways were involved in PLCε-mediated Twist1 expression. The stability of Twist1 was determined after blocking protein synthesis with cycloheximide. Reporter assays utilized E-cadherin or N-cadherin luciferase reporters under depletion of PLCε or Twist1. Transwell assays assessed cell migration. Finally, a nude mouse tumor xenograft assay was conducted to verify the role of PLCε in tumor formation. Results Our findings revealed that the expression of PLCε was positively associated with Twist1 in clinical PCa samples. PLCε knockdown promoted mitochondrial oxidative metabolism in PCa cells. Mechanistically, PLCε increased phosphorylation of Twist1 and stabilized the Twist1 protein through MAPK signaling. The transcriptional activity of Twist1, and the Twist1-mediated epithelial-to-mesenchymal transition, cell migration, and transcription regulation, were suppressed by PLCε knockdown and by blocking PPARβ nuclear translocation. The tumor xenograft assay demonstrated that PLCε depletion diminished PCa cell tumorigenesis. Conclusions These findings reveal an undiscovered physiological role for PLCε in the suppression of mitochondrial oxidative metabolism that has significant implications for understanding PCa occurrence and migration. Electronic supplementary material The online version of this article (10.1186/s13046-019-1323-8) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
de la Cruz López KG, Toledo Guzmán ME, Sánchez EO, García Carrancá A. mTORC1 as a Regulator of Mitochondrial Functions and a Therapeutic Target in Cancer. Front Oncol 2019; 9:1373. [PMID: 31921637 PMCID: PMC6923780 DOI: 10.3389/fonc.2019.01373] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 11/21/2019] [Indexed: 02/05/2023] Open
Abstract
Continuous proliferation of tumor cells requires constant adaptations of energy metabolism to rapidly fuel cell growth and division. This energetic adaptation often comprises deregulated glucose uptake and lactate production in the presence of oxygen, a process known as the "Warburg effect." For many years it was thought that the Warburg effect was a result of mitochondrial damage, however, unlike this proposal tumor cell mitochondria maintain their functionality, and is essential for integrating a variety of signals and adapting the metabolic activity of the tumor cell. The mammalian/mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of numerous cellular processes implicated in proliferation, metabolism, and cell growth. mTORC1 controls cellular metabolism mainly by regulating the translation and transcription of metabolic genes, such as peroxisome proliferator activated receptor γ coactivator-1 α (PGC-1α), sterol regulatory element-binding protein 1/2 (SREBP1/2), and hypoxia inducible factor-1 α (HIF-1α). Interestingly it has been shown that mTORC1 regulates mitochondrial metabolism, thus representing an important regulator in mitochondrial function. Here we present an overview on the role of mTORC1 in the regulation of mitochondrial functions in cancer, considering new evidences showing that mTORC1 regulates the translation of nucleus-encoded mitochondrial mRNAs that result in an increased ATP mitochondrial production. Moreover, we discuss the relationship between mTORC1 and glutaminolysis, as well as mitochondrial metabolites. In addition, mitochondrial fission processes regulated by mTORC1 and its impact on cancer are discussed. Finally, we also review the therapeutic efficacy of mTORC1 inhibitors in cancer treatments, considering its use in combination with other drugs, with particular focus on cellular metabolism inhibitors, that could help improve their anti neoplastic effect and eliminate cancer cells in patients.
Collapse
Affiliation(s)
- Karen Griselda de la Cruz López
- Posgrado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Alejandro García Carrancá
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México & Instituto Nacional de Cancerología, Secretaría de Salud, Mexico City, Mexico
- *Correspondence: Alejandro García Carrancá
| |
Collapse
|