1
|
Promila L, Sarkar K, Guleria S, Rakshit A, Rathore M, Singh NC, Khan S, Tomar MS, Ammanathan V, Barthwal MK, Kumaravelu J, Shrivastava A, Mitra K, Guha R, Aggarwal A, Lahiri A. Mitochondrial calcium uniporter regulates human fibroblast-like synoviocytes invasion via altering mitochondrial dynamics and dictates rheumatoid arthritis pathogenesis. Free Radic Biol Med 2025; 234:55-71. [PMID: 40188890 DOI: 10.1016/j.freeradbiomed.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease that currently has no cure. Fibroblast-like synoviocytes (FLS), present in the RA synovium, play a pivotal role in RA pathogenesis. Notably, FLS in the RA patients (RA-FLS) exhibit characteristics similar to cancer cells, like enhanced migration, invasiveness, uncontrolled proliferation, resistance to apoptosis, and metabolic reprogramming. RA-FLS invasiveness is linked to radiographic joint damage in the patients, whereas inhibiting the FLS migration mitigates disease pathology. However, the molecular mechanisms underlying the migration and invasion capabilities of RA-FLS are not entirely understood. In this work, we have explored the function of mitochondrial calcium uniporter (MCU) and calcium signaling in FLS invasion. Our findings demonstrate a positive correlation between MCU expression and RA disease score. Interestingly, mitochondrial size was reduced, and peripheral localization was more pronounced in the RA-FLS when compared to the control FLS. Mitochondrial calcium import inhibition in the FLS by specific MCU inhibitor, Ruthenium-360 restored these altered mitochondrial dynamics and reduced the invasive phenotype. Through unbiased transcriptome analysis, we identified that MCU-mediated calcium signaling in RA-FLS leads to the enriched actin cytoskeleton and focal adhesion pathways responsible for the invasion phenotype, which can be effectively suppressed by inhibiting MCU. Additionally, we found that mitochondrial transport facilitator Miro1 binds to MCU in a calcium-dependent manner and regulates MCU-mediated mitochondrial dynamics and RA-FLS invasion. Experiments utilizing mice xenograft model demonstrated that MCU silencing diminishes the migration of RA-FLS toward the sites of inflammation in the immunocompromised SCID mice. Altogether, our findings highlight MCU as a promising therapeutic target to inhibit RA-FLS migration and RA progression.
Collapse
Affiliation(s)
- Lakra Promila
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kabita Sarkar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Shivika Guleria
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Adrija Rakshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Manisha Rathore
- Lab Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nishakumari C Singh
- Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shaziya Khan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | | | - Veena Ammanathan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jagavelu Kumaravelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | | | - Kalyan Mitra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Sophisticated Analytical Instrument Facility and Research Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajdeep Guha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Lab Animal Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Amita Aggarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Amit Lahiri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Du X, Huang S, Lin Z, Chen G, Jiang Y, Zhang H. Organelle-targeted small molecular photosensitizers for enhanced photodynamic therapy: a minireview for recent advances and potential applications. Chem Commun (Camb) 2025; 61:7236-7252. [PMID: 40289926 DOI: 10.1039/d5cc01642h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Photodynamic therapy (PDT) is a promising approach for cancer treatment that involves the use of photosensitizers to generate reactive oxygen species upon light irradiation, resulting in selective cytotoxicity. To enhance the efficiency of PDT, researchers have developed organelle-targeting photosensitizers that specifically accumulate in critical cellular organelles. This review provides a comprehensive overview of recent advancements in the development of organelle-targeting photosensitizers for PDT. Different organelles, including mitochondria, plasma membrane, lysosome, endoplasmic reticulum, lipid droplets, nucleus, and Golgi, have been targeted to improve the selectivity and effectiveness of PDT. Various strategies have been employed to design and synthesize these photosensitizers, optimizing their organelle-specific accumulation and photodynamic efficiency. This review discusses the principles and mechanisms underlying the design of organelle-targeting photosensitizers, along with their exceptional results achieved in preclinical studies. Furthermore, potential applications and challenges in the development of multi-organelles-targeting photosensitizers and the synergistic use of multiple photosensitizers targeting different organelles are highlighted. Overall, organelle-targeting photosensitizers offer a promising avenue for advancing the field of PDT and improving its clinical applicability.
Collapse
Affiliation(s)
- Xiaomeng Du
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Shumei Huang
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
- Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Zhenxin Lin
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Gang Chen
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Yin Jiang
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Huatang Zhang
- School of Biomedical and Pharmaceutical Sciences and School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
3
|
Pathak T, Benson JC, Tang PW, Trebak M, Hempel N. Crosstalk between calcium and reactive oxygen species signaling in cancer revisited. Cell Calcium 2025; 127:103014. [PMID: 40139005 DOI: 10.1016/j.ceca.2025.103014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
The homeostasis of cellular reactive oxygen species (ROS) and calcium (Ca2+) are intricately linked. ROS signaling and Ca2+ signaling are reciprocally regulated within cellular microdomains and are crucial for transcription, metabolism and cell function. Tumor cells often highjack ROS and Ca2+ signaling mechanisms to ensure optimal cell survival and tumor progression. Expression and regulation of Ca2+ channels and transporters at the plasma membrane, endoplasmic reticulum, mitochondria and other endomembranes are often altered in tumor cells, and this includes their regulation by ROS and reactive nitrogen species (RNS). Likewise, alterations in cellular Ca2+ levels influence the generation and scavenging of oxidants and thus can alter the redox homeostasis of the cell. This interplay can be either beneficial or detrimental to the cell depending on the localization, duration and levels of ROS and Ca2+ signals. At one end of the spectrum, Ca2+ and ROS/RNS can function as signaling modules while at the other end, lethal surges in these species are associated with cell death. Here, we highlight the interplay between Ca2+ and ROS in cancer progression, emphasize the impact of redox regulation on Ca2+ transport mechanisms, and describe how Ca2+ signaling pathways, in turn, can regulate the cellular redox environment.
Collapse
Affiliation(s)
- Trayambak Pathak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Cory Benson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Priscilla W Tang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, Division of Malignant Hematology & Medical Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Nadine Hempel
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, Division of Malignant Hematology & Medical Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Weissenrieder JS, Peura J, Paudel U, Bhalerao N, Weinmann N, Johnson C, Wengyn M, Drager R, Furth EE, Simin K, Ruscetti M, Stanger BZ, Rustgi AK, Pitarresi JR, Foskett JK. Mitochondrial Ca 2+ controls pancreatic cancer growth and metastasis by regulating epithelial cell plasticity. Cell Rep 2025; 44:115627. [PMID: 40286270 DOI: 10.1016/j.celrep.2025.115627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/24/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Endoplasmic reticulum to mitochondria Ca2+ transfer is important for cancer cell survival, but the role of mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU) in pancreatic ductal adenocarcinoma (PDAC) is poorly understood. Here, we show that increased MCU expression is associated with malignancy and poorer outcomes in patients with PDAC. In isogenic murine PDAC models, Mcu deletion (McuKO) ablated mitochondrial Ca2+ uptake, which reduced proliferation and inhibited self-renewal. Orthotopic implantation of MCU-null tumor cells reduced primary tumor growth and metastasis. Mcu deletion reduced the cellular plasticity of tumor cells by inhibiting epithelial-to-mesenchymal transition (EMT), which contributes to metastatic competency in PDAC. Mechanistically, the loss of mitochondrial Ca2+ uptake reduced the expression of the key EMT transcription factor Snail and secretion of the EMT-inducing ligand TGF-β. Snail re-expression and TGF-β treatment rescued deficits in McuKO cells and restored their metastatic ability. Thus, MCU may present a therapeutic target in PDAC to limit cancer-cell-induced EMT and metastasis.
Collapse
Affiliation(s)
- Jillian S Weissenrieder
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jessica Peura
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Usha Paudel
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nikita Bhalerao
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Natalie Weinmann
- Department of Chemistry, Millersville University, Millersville, PA, USA
| | - Calvin Johnson
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Maximilian Wengyn
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5157, USA
| | - Rebecca Drager
- Department of Chemistry, The Ohio State University, Columbus, OH, USA
| | - Emma Elizabeth Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Karl Simin
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ben Z Stanger
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5157, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York City, NY 10032, USA
| | - Jason R Pitarresi
- Division of Hematology/Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - J Kevin Foskett
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Arcos M, Goodla L, Kim H, Desai SP, Liu R, Yin K, Liu Z, Martin DR, Xue X. PINK1-deficiency facilitates mitochondrial iron accumulation and colon tumorigenesis. Autophagy 2025; 21:737-753. [PMID: 39512202 DOI: 10.1080/15548627.2024.2425594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/25/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
Mitophagy, the process by which cells eliminate damaged mitochondria, is mediated by PINK1 (PTEN induced kinase 1). Our recent research indicates that PINK1 functions as a tumor suppressor in colorectal cancer by regulating cellular metabolism. Interestingly, PINK1 ablation activated the NLRP3 (NLR family pyrin domain containing 3) inflammasome, releasing IL1B (interleukin 1 beta). However, inhibiting the NLRP3-IL1B signaling pathway with an IL1R (interleukin 1 receptor) antagonist or NLRP3 inhibitor did not hinder colon tumor growth after PINK1 loss. To identify druggable targets in PINK1-deficient tumors, ribonucleic acid sequencing analysis was performed on colon tumors from pink1 knockout and wild-type mice. Gene Set Enrichment Analysis highlighted the enrichment of iron ion transmembrane transporter activity. Subsequent qualitative polymerase chain reaction and western blot analysis revealed an increase in mitochondrial iron transporters, including mitochondrial calcium uniporter, in PINK1-deficient colon tumor cells and tissues. Live-cell iron staining demonstrated elevated cellular and mitochondrial iron levels in PINK1-deficient cells. Clinically used drugs deferiprone and minocycline reduced mitochondrial iron and superoxide levels, resulting in decreased colon tumor cell growth in vitro and in vivo. Manipulating the mitochondrial iron uptake protein MCU (mitochondrial calcium uniporter) also affected cell and xenograft tumor growth. This study suggests that therapies aimed at reducing mitochondrial iron levels may effectively inhibit colon tumor growth, particularly in patients with low PINK1 expression.Abbreviation: ANOVA: analysis of variance; APC: adenomatous polyposis coli; cAMP: cyclic adenosine monophosphate; CDX2: caudal type homeobox 2; CGAS: cyclic GMP-AMP synthase; CRC: colorectal cancer; DNA: deoxyribonucleic acid; DFP: deferiprone; DMEM: Dulbecco's modified Eagle medium; DSS: dextran sodium sulfate; ERT2-Cre: Cre recombinase fused to a triple mutant form of the human estrogen receptor; EV: empty vector; GLB: glybenclamide/glyburide; H&E: hematoxylin and eosin; ICP-MS: inductively coupled plasma mass spectrometer; IL1B: interleukin 1 beta; kDa: kilodalton; MCU: mitochondrial calcium uniporter; MKI67: marker of proliferation Ki-67; mRNA: messenger ribonucleic acid; MTT: 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide; NLRP3: NLR family pyrin domain containing 3; OE: overexpression; PBS: phosphate-buffered saline; p-CREB: phosphorylated cAMP responsive element binding protein; PINK1: PTEN induced kinase 1; p-PRKAA/AMPK: phosphorylated protein kinase AMP-activated catalytic subunit alpha; qPCR: qualitative polymerase chain reaction; RNA-seq: ribonucleic acid sequencing; ROS: reactive oxygen species; sg: single guide; sh: short hairpin; SLC25A28: solute carrier family 25 member 28; SLC25A37/MFRN: solute carrier family 25 member 37; STING1: stimulator of interferon response cGAMP interactor 1; TP53/p53: tumor protein p53; TUBA: tubulin alpha; µL: microliter; µm: micrometer; µM: micromolar; mm: millimeter.
Collapse
Affiliation(s)
- Mariella Arcos
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Lavanya Goodla
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Hyeoncheol Kim
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Sharina P Desai
- Department of Molecular Genetics Microbiology, University of New Mexico, Albuquerque, NM, USA
| | - Rui Liu
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, NM, USA
| | - Kunlun Yin
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - Zhaoli Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| | - David R Martin
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Xiang Xue
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
6
|
Lin HY, Chu PY. Mitochondrial calcium uniporter as biomarker and therapeutic target for breast cancer: Prognostication, immune microenvironment, epigenetic regulation and precision medicine. J Adv Res 2025; 70:445-461. [PMID: 38663838 PMCID: PMC11976406 DOI: 10.1016/j.jare.2024.04.015] [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: 12/12/2023] [Revised: 03/24/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Mitochondrial calcium uniporter (MCU) is a central subunit of MCU complex that regulate the levels of calcium ions within mitochondria. A comprehensive understanding the implications of MCU in clinical prognostication, biological understandings and therapeutic opportunity of breast cancer (BC) is yet to be determined. OBJECTIVES This study aims to investigate the role of MCU in predictive performance, tumor progression, epigenetic regulation, shaping of tumor immune microenvironment, and pharmacogenetics and the development of anti-tumor therapy for BC. METHODS The downloaded TCGA datasets were used to identify predictive ability of MCU expressions via supervised learning principle. Functional enrichment, mutation landscape, immunological profile, drug sensitivity were examined using bioinformatics analysis and confirmed by experiments exploiting human specimens, in vitro and in vivo models. RESULTS MCU copy numbers increase with MCU gene expression. MCU expression, but not MCU genetic alterations, had a positive correlation with known BC prognostic markers. Higher MCU levels in BC showed modest efficacy in predicting overall survival. In addition, high MCU expression was associated with known BC prognostic markers and with malignancy. In BC tumor and sgRNA-treated cell lines, enrichment pathways identified the involvement of cell cycle and immunity. miR-29a was recognized as a negative epigenetic regulator of MCU. High MCU levels were associated with increased mutation levels in oncogene TP53 and tumor suppression gene CDH1, as well as with an immunosuppressive microenvironment. Sigle-cell sequencing indicated that MCU mostly mapped on to tumor cell and CD8 T-cells. Inter-databases verification further confirmed the aforementioned observation. miR-29a-mediated knockdown of MCU resulted in tumor suppression and mitochondrial dysfunction, as well as diminished metastasis. Furthermore, MCU present pharmacogenetic significance in cellular docetaxel sensitivity and in prediction of patients' response to chemotherapeutic regimen. CONCLUSION MCU shows significant implication in prognosis, outcome prediction, microenvironmental shaping and precision medicine for BC. miR-29a-mediated MCU inhibition exerts therapeutic effect in tumor growth and metastasis.
Collapse
Affiliation(s)
- Hung-Yu Lin
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Pei-Yi Chu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan; Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
7
|
Wang J, Jiang J, Hu H, Chen L. MCU complex: Exploring emerging targets and mechanisms of mitochondrial physiology and pathology. J Adv Res 2025; 68:271-298. [PMID: 38417574 PMCID: PMC11785567 DOI: 10.1016/j.jare.2024.02.013] [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: 11/16/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Globally, the onset and progression of multiple human diseases are associated with mitochondrial dysfunction and dysregulation of Ca2+ uptake dynamics mediated by the mitochondrial calcium uniporter (MCU) complex, which plays a key role in mitochondrial dysfunction. Despite relevant studies, the underlying pathophysiological mechanisms have not yet been fully elucidated. AIM OF REVIEW This article provides an in-depth analysis of the current research status of the MCU complex, focusing on its molecular composition, regulatory mechanisms, and association with diseases. In addition, we conducted an in-depth analysis of the regulatory effects of agonists, inhibitors, and traditional Chinese medicine (TCM) monomers on the MCU complex and their application prospects in disease treatment. From the perspective of medicinal chemistry, we conducted an in-depth analysis of the structure-activity relationship between these small molecules and MCU and deduced potential pharmacophores and binding pockets. Simultaneously, key structural domains of the MCU complex in Homo sapiens were identified. We also studied the functional expression of the MCU complex in Drosophila, Zebrafish, and Caenorhabditis elegans. These analyses provide a basis for exploring potential treatment strategies targeting the MCU complex and provide strong support for the development of future precision medicine and treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW The MCU complex exhibits varying behavior across different tissues and plays various roles in metabolic functions. It consists of six MCU subunits, an essential MCU regulator (EMRE), and solute carrier 25A23 (SLC25A23). They regulate processes, such as mitochondrial Ca2+ (mCa2+) uptake, mitochondrial adenosine triphosphate (ATP) production, calcium dynamics, oxidative stress (OS), and cell death. Regulation makes it a potential target for treating diseases, especially cardiovascular diseases, neurodegenerative diseases, inflammatory diseases, metabolic diseases, and tumors.
Collapse
Affiliation(s)
- Jin Wang
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China
| | - Jinyong Jiang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou 416000, China
| | - Haoliang Hu
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China; College of Medicine, Hunan University of Arts and Science, Changde 415000, China.
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Learning Key Laboratory for Pharmacoproteomics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang 421001, China.
| |
Collapse
|
8
|
Cartes-Saavedra B, Ghosh A, Hajnóczky G. The roles of mitochondria in global and local intracellular calcium signalling. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00820-1. [PMID: 39870977 DOI: 10.1038/s41580-024-00820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/29/2025]
Abstract
Activation of Ca2+ channels in Ca2+ stores in organelles and the plasma membrane generates cytoplasmic calcium ([Ca2+]c) signals that control almost every aspect of cell function, including metabolism, vesicle fusion and contraction. Mitochondria have a high capacity for Ca2+ uptake and chelation, alongside efficient Ca2+ release mechanisms. Still, mitochondria do not store Ca2+ in a prolonged manner under physiological conditions and lack the capacity to generate global [Ca2+]c signals. However, mitochondria take up Ca2+ at high local [Ca2+]c signals that originate from neighbouring organelles, and also during sustained global elevations of [Ca2+]c. Accumulated Ca2+ in the mitochondria stimulates oxidative metabolism and upon return to the cytoplasm, can produce spatially confined rises in [Ca2+]c to exert control over processes that are sensitive to Ca2+. Thus, the mitochondrial handling of [Ca2+]c is of physiological relevance. Furthermore, dysregulation of mitochondrial Ca2+ handling can contribute to debilitating diseases. We discuss the mechanisms and relevance of mitochondria in local and global calcium signals.
Collapse
Affiliation(s)
- Benjamín Cartes-Saavedra
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Arijita Ghosh
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
SO EDMUNDCHEUNG, CHOW LOUISWC, CHUANG CHINMIN, CHEN CINGYU, WU CHENGHSUN, SHIAO LIANRU, OU TINGTSZ, WONG KARLOK, LEUNG YUKMAN, HUANG YIPING. MCU-i4, a mitochondrial Ca 2+ uniporter modulator, induces breast cancer BT474 cell death by enhancing glycolysis, ATP production and reactive oxygen species (ROS) burst. Oncol Res 2025; 33:397-406. [PMID: 39866241 PMCID: PMC11753992 DOI: 10.32604/or.2024.052743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/29/2024] [Indexed: 01/28/2025] Open
Abstract
Objectives Mitochondrial Ca2+ uniporter (MCU) provides a Ca2+ influx pathway from the cytosol into the mitochondrial matrix and a moderate mitochondrial Ca2+ rise stimulates ATP production and cell growth. MCU is highly expressed in various cancer cells including breast cancer cells, thereby increasing the capacity of mitochondrial Ca2+ uptake, ATP production, and cancer cell proliferation. The objective of this study was to examine MCU inhibition as an anti-cancer mechanism. Methods The effects of MCU-i4, a newly developed MCU inhibitor, on cell viability, apoptosis, cytosolic Ca2+, mitochondrial Ca2+ and potential, glycolytic rate, generation of ATP, and reactive oxygen species, were examined in breast cancer BT474 cells. Results MCU-i4 caused apoptotic cell death, and it decreased and increased, respectively, mitochondrial and cytosolic Ca2+ concentration. Inhibition of MCU by MCU-i4 revealed that cytosolic Ca2+ elevation resulted from endoplasmic reticulum (ER) Ca2+ release via inositol 1,4,5-trisphosphate receptors (IP3R) and ryanodine receptors (RYR). Unexpectedly, MCU-i4 enhanced glycolysis and ATP production; it also triggered a large production of reactive oxygen species (ROS) and mitochondrial membrane potential collapse. Conclusion Cytotoxic mechanisms of MCU-i4 in cancer cells involved enhanced glycolysis and heightened formation of ATP and ROS. It is conventionally believed that cancer cell death could be caused by inhibition of glycolysis. Our observations suggest cancer cell death could also be induced by increased glycolytic metabolism.
Collapse
Affiliation(s)
- EDMUND CHEUNG SO
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan, 709, Taiwan
- Graduate Institute of Medical Sciences, Chang Jung Christian University, Tainan, 711301, Taiwan
| | | | - CHIN-MIN CHUANG
- Department of Emergency Medicine, China Medical University Hospital, Taichung, 404327, Taiwan
| | - CING YU CHEN
- Department of Cosmetic Science, Providence University, Taichung, 43301, Taiwan
- School of Pharmacy, China Medical University, Taichung, 404328, Taiwan
| | - CHENG-HSUN WU
- Department of Anatomy, China Medical University, Taichung, 404328, Taiwan
| | - LIAN-RU SHIAO
- Department of Physiology, China Medical University, Taichung, 404328, Taiwan
| | - TING-TSZ OU
- Department of Medicinal Botanicals and Healthcare, Dayeh University, Changhua, 51591, Taiwan
| | - KAR-LOK WONG
- Department of Anesthesiology, Kuang Tien General Hospital, Shalu, Taichung, 433, Taiwan
| | - YUK-MAN LEUNG
- Department of Physiology, China Medical University, Taichung, 404328, Taiwan
| | - YI-PING HUANG
- Department of Physiology, China Medical University, Taichung, 404328, Taiwan
| |
Collapse
|
10
|
Priya, Kumar A, Kumar D. Molecular heterogeneity and MYC dysregulation in triple-negative breast cancer: genomic advances and therapeutic implications. 3 Biotech 2025; 15:33. [PMID: 39777154 PMCID: PMC11700964 DOI: 10.1007/s13205-024-04195-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a diverse range of molecular features that have been extensively studied. MYC plays a critical role in regulating metabolism, differentiation, proliferation, cell growth, and apoptosis. Dysregulation of MYC is associated with poor prognosis and contributes to the development and progression of breast cancer. A particularly intriguing aspect of TNBC is its association with tumors in BRCA1 mutation carriers, especially in younger women. MYC may also contribute to resistance to adjuvant treatments. For TNBC, targeting MYC-regulated pathways in combination with inhibitors of other carcinogenic pathways offers a promising therapeutic approach. Several signaling pathways regulate TNBC, and targeting these pathways could lead to effective therapeutic strategies for breast cancer. Advances in genomic tools, such as CRISPR-Cas9, next-generation sequencing, and whole-exome sequencing, are revolutionizing breast cancer diagnoses. These technologies have significantly enhanced our understanding of MYC oncogenesis, particularly through CRISPR-Cas9 and NGS. Targeting MYC and its partner MAX could provide valuable insights into TNBC. Moreover, the therapeutic potential of targeting MYC-driven signaling mechanisms and their interactions with other oncogenic pathways, including PI3K/AKT/mTOR and Wnt/β-catenin, is increasingly recognized. Next-generation sequencing and CRISPR-Cas9 represent significant breakthroughs in genomic tools that open new opportunities to explore MYC's role in TNBC and facilitate the development of personalized treatment plans. This review discusses the future clinical applications of personalized treatment strategies for patients with TNBC.
Collapse
Affiliation(s)
- Priya
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| | - Arun Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar 801505 India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| |
Collapse
|
11
|
Weng Y, Wang Z, Sitosari H, Ono M, Okamura H, Oohashi T. O-GlcNAcylation regulates osteoblast differentiation through the morphological changes in mitochondria, cytoskeleton, and endoplasmic reticulum. Biofactors 2025; 51:e2131. [PMID: 39405562 DOI: 10.1002/biof.2131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/01/2024] [Indexed: 12/29/2024]
Abstract
To explore the potential mechanisms which O-linked-N-acetylglucosaminylation (O-GlcNAcylation) regulates osteogenesis, a publicly RNA-seq dataset was re-analyzed with literature-mining and showed the primary targets of O-GlcNAcylation in osteoblasts are mitochondria/cytoskeleton. Although the O-GlcNAcylation-regulated mitochondria/cytoskeleton has been extensively studied, its specific role during osteogenesis remains unclear. To address this, we knocked out Ogt (Ogt-KO) in MC3T3-E1 osteoblastic cells. Then, significantly reduced osteoblast differentiation, motility, proliferation, mitochondria-endoplasmic reticulum (Mito-ER) coupling, volume of ER, nuclear tubulins, and oxygen metabolism were observed in Ogt-KO cells. Through artificial intelligence (AI)-predicted cellular structures, the time-lapse live cells imaging with reactive-oxygen-species/hypoxia staining showed that lower cell proliferation and altered oxygen metabolism in the Ogt-KO cells were correlated with the Mito-ER coupling. Bioinformatics analysis, combined with correlated mRNA and protein expression, suggested that Ezh2 and its downstream targets (Opa1, Gsk3a, Wnt3a, Hif1a, and Hspa9) may be involved in O-GlcNAcylation-regulated Mito-ER coupling, ultimately impacting osteoblast differentiation. In conclusion, our findings indicate that O-GlcNAcylation-regulated osteoblast differentiation is linked to morphological changes in mitochondria, cytoskeleton, and ER, with Ezh2 potentially playing a crucial role.
Collapse
Affiliation(s)
- Yao Weng
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Heriati Sitosari
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Department of Oral Biology, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mitsuaki Ono
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama, Japan
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
12
|
Shumanska M, Lodygin D, Gibhardt CS, Ickes C, Stejerean-Todoran I, Krause LCM, Pahl K, Jacobs LJHC, Paluschkiwitz A, Liu S, Boshnakovska A, Voigt N, Legler TJ, Haubrock M, Mitkovski M, Poschmann G, Rehling P, Dennerlein S, Riemer J, Flügel A, Bogeski I. Mitochondrial calcium uniporter complex controls T-cell-mediated immune responses. EMBO Rep 2025; 26:407-442. [PMID: 39623165 PMCID: PMC11772621 DOI: 10.1038/s44319-024-00313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 01/29/2025] Open
Abstract
T-cell receptor (TCR)-induced Ca2+ signals are essential for T-cell activation and function. In this context, mitochondria play an important role and take up Ca2+ to support elevated bioenergetic demands. However, the functional relevance of the mitochondrial-Ca2+-uniporter (MCU) complex in T-cells was not fully understood. Here, we demonstrate that TCR activation causes rapid mitochondrial Ca2+ (mCa2+) uptake in primary naive and effector human CD4+ T-cells. Compared to naive T-cells, effector T-cells display elevated mCa2+ and increased bioenergetic and metabolic output. Transcriptome and proteome analyses reveal molecular determinants involved in the TCR-induced functional reprogramming and identify signalling pathways and cellular functions regulated by MCU. Knockdown of MCUa (MCUaKD), diminishes mCa2+ uptake, mitochondrial respiration and ATP production, as well as T-cell migration and cytokine secretion. Moreover, MCUaKD in rat CD4+ T-cells suppresses autoimmune responses in an experimental autoimmune encephalomyelitis (EAE) multiple sclerosis model. In summary, we demonstrate that mCa2+ uptake through MCU is essential for proper T-cell function and has a crucial role in autoimmunity. T-cell specific MCU inhibition is thus a potential tool for targeting autoimmune disorders.
Collapse
Affiliation(s)
- Magdalena Shumanska
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Dmitri Lodygin
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Christian Ickes
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Ioana Stejerean-Todoran
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Lena C M Krause
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Kira Pahl
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Lianne J H C Jacobs
- Redox Metabolism, Institute of Biochemistry and CECAD, University of Cologne, Cologne, Germany
| | - Andrea Paluschkiwitz
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Shuya Liu
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Angela Boshnakovska
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Centre, Göttingen, Germany
| | - Martin Haubrock
- Department of Medical Bioinformatics, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gereon Poschmann
- Institute for Molecular Medicine, Proteome Research, University Hospital and Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Rehling
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Sven Dennerlein
- Department of Cellular Biochemistry, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Jan Riemer
- Redox Metabolism, Institute of Biochemistry and CECAD, University of Cologne, Cologne, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Centre, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
13
|
Bustos G, Ahumada-Castro U, Silva-Pavez E, Huerta H, Puebla A, Quezada C, Morgado-Cáceres P, Casanova-Canelo C, Smith-Cortinez N, Podunavac M, Oyarce C, Lladser A, Farias P, Lovy A, Molgó J, Torres VA, Zakarian A, Cárdenas JC. The IP 3R inhibitor desmethylxestospongin B reduces tumor cell migration, invasion and metastasis by impairing lysosome acidification and β1-integrin recycling. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167557. [PMID: 39486657 DOI: 10.1016/j.bbadis.2024.167557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Cancer is the second leading cause of death worldwide. >90 % of cancer-related deaths are due to metastasis, a process that depends on the ability of cancer cells to leave the primary tumor, migrate, and colonize different tissues. Inositol 1,4,5-trisphosphate receptor (IP3R)-mediated Ca2+ signaling plays an essential role in maintaining the homeostasis of cancer cells and the sustained proliferation. Desmethylxestospongin B (dmXeB) is a specific inhibitor of the IP3R that selectively arrests cell proliferation and promotes cancer cell death at high concentrations. However, whether migration, invasion and metastasis can be affected by this drug is unknown. Here, by using the highly metastatic triple negative breast cancer (TNBC) cell line MDA-MB-231, we demonstrate that a prolonged inhibition of IP3R-mediated Ca2+ signals with dmXeB significantly reduces cell migration and invasion in vitro and metastasis in vivo. We found that this phenomenon was independent of the bioenergetic control of IP3R over the mitochondria and AMPK activation. Furthermore, employing a tandem LC3-GFP-mcherry assay, we found that prolonged inhibition of IP3R with dmXeB leads to diminished autophagic flux. This reduction can be attributed to impaired lysosomal acidification, as evidenced by assessments using DQ-BSA and pHrodo. Since cell migration requires appropriate assembly and disassembly of focal adhesions, along with the internalization and recycling of integrins via autophagy, we explored the dependency of integrin recycling from autophagosomes, finding that IP3R inhibition with dmXeB impaired the recycling of β1-integrins, which accumulated within autophagosomes. Our findings reveal an unexpected effect of IP3R inhibition with dmXeB in cancer cells that could represent a novel therapeutic strategy for the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Galdo Bustos
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Ulises Ahumada-Castro
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Eduardo Silva-Pavez
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile
| | - Hernán Huerta
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Andrea Puebla
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Camila Quezada
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Pablo Morgado-Cáceres
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - César Casanova-Canelo
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Natalia Smith-Cortinez
- Department of Gastroenterology and Hepatology, UMCG, University of Groningen, Netherlands
| | - Maša Podunavac
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Cesar Oyarce
- Department of Medical Microbiology and Infection Prevention, Tumor Virology and Cancer Immunotherapy, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alvaro Lladser
- Centro Cientifico y Tecnologico de Excelencia Ciencia & Vida, Fundación Ciencia and Vida, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Paula Farias
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Alenka Lovy
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile
| | - Jordi Molgó
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé, Service d'Ingénierie Moléculaire pour la Santé (SIMoS), Equipe Mixte de Recherche CNRS 9004, F-91191 Gif-sur-Yvette, France
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Chile; Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380453, Chile; Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Armen Zakarian
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - J César Cárdenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile; Geroscience Center for Brain Health and Metabolism, Santiago 8580745, Chile; Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA; The Buck Institute for Research on Aging, Novato, USA.
| |
Collapse
|
14
|
Zhang X, Zhang H, Dong Q, Qin Y, Cao Y, Zhu H, Ma Z, Li Z, Rao Z, Ning P, Tian Z, Xia Y, Yang P, Wang Z. Bypassing Ca 2+ Influx for Antimetastasis Photodynamic Therapy via Robust Nucleus-Targeted Near-Infrared Cyanines. NANO LETTERS 2024; 24:15817-15826. [PMID: 39584561 DOI: 10.1021/acs.nanolett.4c04789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Hypoxia-induced tumor metastasis severely hinders the efficacy of photodynamic therapy (PDT) in cancer treatment. Current strategies predominantly offer palliative suppression of the HIF-1α pathway, emphasizing the urgent need for innovative PDT approaches to prevent metastasis from the outset. Our study revealed that typical PDT triggers an increase in cytoplasmic Ca2+ levels, activating HIF-1α, and that reducing Ca2+ levels can, in turn, mitigate metastasis. Considering cytoplasm's role in Ca2+ storage and regulation, we propose that PDT-induced metastasis can be addressed at its source by precise intracellular localization of photosensitizers (PSs). We developed near-infrared (NIR) cyanine PSs with inherent nucleus targeting capabilities. These PSs effectively inhibit cytoplasmic Ca2+ elevation and reduce HIF-1α activity upon irradiation, achieving remarkable antimetastatic effects in 4T1 tumors. Consequently, our findings highlight the pivotal role of Ca2+ in PDT-induced metastasis and provide a robust approach for circumventing metastasis from the outset using new nucleus-targeting organic PSs.
Collapse
Affiliation(s)
- Xianghan Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Huaicong Zhang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou, Guangdong 510555, China
| | - Qunyan Dong
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yuan Qin
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Yutian Cao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Haixing Zhu
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zimeng Ma
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zehua Li
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhiping Rao
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Pengbo Ning
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zuhong Tian
- State Key Laboratory of Cancer Biology & XiJing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Yuqiong Xia
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Peng Yang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular and Neuro Imaging (Ministry of Education), School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| |
Collapse
|
15
|
Chen F, Xue Y, Zhang W, Zhou H, Zhou Z, Chen T, YinWang E, Li H, Ye Z, Gao J, Wang S. The role of mitochondria in tumor metastasis and advances in mitochondria-targeted cancer therapy. Cancer Metastasis Rev 2024; 43:1419-1443. [PMID: 39307891 PMCID: PMC11554835 DOI: 10.1007/s10555-024-10211-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/08/2024] [Indexed: 11/05/2024]
Abstract
Mitochondria are central actors in diverse physiological phenomena ranging from energy metabolism to stress signaling and immune modulation. Accumulating scientific evidence points to the critical involvement of specific mitochondrial-associated events, including mitochondrial quality control, intercellular mitochondrial transfer, and mitochondrial genetics, in potentiating the metastatic cascade of neoplastic cells. Furthermore, numerous recent studies have consistently emphasized the highly significant role mitochondria play in coordinating the regulation of tumor-infiltrating immune cells and immunotherapeutic interventions. This review provides a comprehensive and rigorous scholarly investigation of this subject matter, exploring the intricate mechanisms by which mitochondria contribute to tumor metastasis and examining the progress of mitochondria-targeted cancer therapies.
Collapse
Affiliation(s)
- Fanglu Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yucheng Xue
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenkan Zhang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hao Zhou
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhiyi Zhou
- The First People's Hospital of Yuhang District, Hangzhou, Zhejiang, China
| | - Tao Chen
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Eloy YinWang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Hengyuan Li
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhaoming Ye
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Shengdong Wang
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P.R. China.
- Institute of Orthopedic Research, Zhejiang University, Hangzhou, 310009, P.R. China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
16
|
Stejerean-Todoran I, Gibhardt CS, Bogeski I. Calcium signals as regulators of ferroptosis in cancer. Cell Calcium 2024; 124:102966. [PMID: 39504596 DOI: 10.1016/j.ceca.2024.102966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
The field of ferroptosis research has grown exponentially since this form of cell death was first identified over a decade ago. Ferroptosis, an iron- and ROS-dependent type of cell death, is controlled by various metabolic pathways, including but not limited to redox and calcium (Ca2+) homeostasis, iron fluxes, mitochondrial function and lipid metabolism. Importantly, therapy-resistant tumors are particularly susceptible to ferroptotic cell death, rendering ferroptosis a promising therapeutic strategy against numerous malignancies. Calcium signals are important regulators of both cancer progression and cell death, with recent studies indicating their involvement in ferroptosis. Cells undergoing ferroptosis are characterized by plasma membrane rupture and the formation of nanopores, which facilitate influx of ions such as Ca2+ into the affected cells. Furthermore, mitochondrial Ca²⁺ levels have been implicated in directly influencing the cellular response to ferroptosis. Despite the remarkable progress made in the field, our understanding of the contribution of Ca2+ signals to ferroptosis remains limited. Here, we summarize key connections between Ca²⁺ signaling and ferroptosis in cancer pathobiology and discuss their potential therapeutic significance.
Collapse
Affiliation(s)
- Ioana Stejerean-Todoran
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Christine S Gibhardt
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Ivan Bogeski
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
17
|
Toussaint A, Singh M, Wang G, Driscoll M, Bhatt V, Ndong JDLC, Shuaib S, Zoltowski H, Gilleran J, Peng Y, Tsymbal A, Jia D, Roberge J, Chiou H, Guo JY, Herranz D, Langenfeld J. BMP receptor 2 inhibition regulates mitochondrial bioenergetics to induce synergistic cell death with BCL-2 inhibitors in leukemia and NSLC cells. RESEARCH SQUARE 2024:rs.3.rs-5065904. [PMID: 39315260 PMCID: PMC11419183 DOI: 10.21203/rs.3.rs-5065904/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Bone morphogenetic protein (BMP) signaling cascade is a phylogenetically conserved stem cell regulator that is aberrantly expressed in non-small cell lung cancer (NSLC) and leukemias. BMP signaling negatively regulates mitochondrial bioenergetics in lung cancer cells. The impact of inhibiting BMP signaling on mitochondrial bioenergetics and the effect this has on the survival of NSLC and leukemia cells are not known. Methods Utilizing the BMP type 2 receptor (BMPR2) JL189, BMPR2 knockout (KO) in cancer cells, and BMP loss of function mutants in C elegans, we determined the effects of BMPR2 inhibition (BMPR2i) on TCA cycle metabolic intermediates, mitochondrial respiration, and the regulation of mitochondrial superoxide anion (SOA) and Ca++ levels. We also examined whether BMPR2i altered the threshold cancer therapeutics induce cell death in NSLC and leukemia cell lines. KO of the mitochondria uniporter (MCU) was used to determine the mechanism BMPR2i regulates the uptake of Ca++ into the mitochondria, mitochondrial bioenergetics, and cell death. Results BMPR2i increases mtCa++ levels and enhances mitochondrial bioenergetics in both NSLC and leukemia cell lines that is conserved in C elegans. BMPR2i induced increase in mtCa++ levels is regulated through the MCU, effecting mitochondria mass and cell survival. BMPR2i synergistically induced cell death when combined with BCL-2 inhibitors or microtubule targeting agents in both NSLC and leukemia cells. Cell death is caused by synergistic increase in mitochondrial ROS and Ca++ levels. BMPR2i enhances Ca++ uptake into the mitochondria induced by reactive oxygen species (ROS) produced by cancer therapeutics. Both acute myeloid leukemia (AML) and T-cell lymphoblastic leukemia cells lines were more responsive to the JL189 alone and when combined with venetoclax or navitoclax compared to NSLC.
Collapse
|
18
|
Zhou C, Cui H, Yang Y, Chen L, Feng M, Gao Y, Li D, Li L, Chen X, Li X, Cao Y. SEC61 translocon gamma subunit is correlated with glycolytic activity, epithelial mesenchymal transition and the immune suppressive phenotype of lung adenocarcinoma. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1748-1760. [PMID: 38978503 PMCID: PMC11693863 DOI: 10.3724/abbs.2024109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/08/2024] [Indexed: 07/10/2024] Open
Abstract
Lung adenocarcinoma (LUAD) remains a predominant cause of cancer-related mortality globally, underscoring the urgency for targeted therapeutic strategies. The specific role and impact of the SEC61 translocon gamma subunit (SEC61G) in LUAD progression and metastasis remain largely unexplored. In this study, we use a multifaceted approach, combining bioinformatics analysis with experimental validation, to elucidate the pivotal role of SEC61G and its associated molecular mechanisms in LUAD. Our integrated analyses reveal a significant positive correlation between SEC61G expression and the glycolytic activity of LUAD, as evidenced by increased fluorodeoxyglucose (FDG) uptake on positron emission tomography (PET)/CT scans. Further investigations show the potential influence of SEC61G on metabolic reprogramming, which contributes to the immunosuppressive tumor microenvironment (TME). Remarkably, we identify a negative association between SEC61G expression levels and the infiltration of critical immune cell populations within the TME, along with correlations with immune checkpoint gene expression and tumor heterogeneity scores in LUAD. Functional studies demonstrate that SEC61G knockdown markedly inhibits the migration of A549 and H2030 LUAD cells. This inhibitory effect is accompanied by a significant downregulation of key regulators of tumor progression, including hypoxia-inducible factor-1 alpha (HIF-1α), lactate dehydrogenase A, and genes involved in the epithelial-mesenchymal transition pathway. In conclusion, our comprehensive analyses position SEC61G as a potential prognostic biomarker intricately linked to glycolytic metabolism, the EMT pathway, and the establishment of an immune-suppressive phenotype in LUAD. These findings underscore the potential of SEC61G as a therapeutic target and predictive marker for immunotherapeutic responses in LUAD patients.
Collapse
Affiliation(s)
- Changshuai Zhou
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Huanhuan Cui
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yuechao Yang
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Lei Chen
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Mingtao Feng
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yang Gao
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Deheng Li
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Liangdong Li
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xin Chen
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xiaoqiu Li
- Department of PathologyFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yiqun Cao
- Department of NeurosurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
19
|
Colussi DM, Stathopulos PB. The mitochondrial calcium uniporter: Balancing tumourigenic and anti-tumourigenic responses. J Physiol 2024; 602:3315-3339. [PMID: 38857425 DOI: 10.1113/jp285515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Increased malignancy and poor treatability associated with solid tumour cancers have commonly been attributed to mitochondrial calcium (Ca2+) dysregulation. The mitochondrial Ca2+ uniporter complex (mtCU) is the predominant mode of Ca2+ uptake into the mitochondrial matrix. The main components of mtCU are the pore-forming mitochondrial Ca2+ uniporter (MCU) subunit, MCU dominant-negative beta (MCUb) subunit, essential MCU regulator (EMRE) and the gatekeeping mitochondrial Ca2+ uptake 1 and 2 (MICU1 and MICU2) proteins. In this review, we describe mtCU-mediated mitochondrial Ca2+ dysregulation in solid tumour cancer types, finding enhanced mtCU activity observed in colorectal cancer, breast cancer, oral squamous cell carcinoma, pancreatic cancer, hepatocellular carcinoma and embryonal rhabdomyosarcoma. By contrast, decreased mtCU activity is associated with melanoma, whereas the nature of mtCU dysregulation remains unclear in glioblastoma. Furthermore, we show that numerous polymorphisms associated with cancer may alter phosphorylation sites on the pore forming MCU and MCUb subunits, which cluster at interfaces with EMRE. We highlight downstream/upstream biomolecular modulators of MCU and MCUb that alter mtCU-mediated mitochondrial Ca2+ uptake and may be used as biomarkers or to aid in the development of novel cancer therapeutics. Additionally, we provide an overview of the current small molecule inhibitors of mtCU that interact with the Asp residue of the critical Asp-Ile-Met-Glu motif or through other allosteric regulatory mechanisms to block Ca2+ permeation. Finally, we describe the relationship between MCU- and MCUb-mediating microRNAs and mitochondrial Ca2+ uptake that should be considered in the discovery of new treatment approaches for cancer.
Collapse
Affiliation(s)
- Danielle M Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
20
|
Mesa D, Barbieri E, Raimondi A, Freddi S, Miloro G, Jendrisek G, Caldieri G, Quarto M, Schiano Lomoriello I, Malabarba MG, Bresci A, Manetti F, Vernuccio F, Abdo H, Scita G, Lanzetti L, Polli D, Tacchetti C, Pinton P, Bonora M, Di Fiore PP, Sigismund S. A tripartite organelle platform links growth factor receptor signaling to mitochondrial metabolism. Nat Commun 2024; 15:5119. [PMID: 38879572 PMCID: PMC11180189 DOI: 10.1038/s41467-024-49543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 06/08/2024] [Indexed: 06/19/2024] Open
Abstract
One open question in the biology of growth factor receptors is how a quantitative input (i.e., ligand concentration) is decoded by the cell to produce specific response(s). Here, we show that an EGFR endocytic mechanism, non-clathrin endocytosis (NCE), which is activated only at high ligand concentrations and targets receptor to degradation, requires a tripartite organelle platform involving the plasma membrane (PM), endoplasmic reticulum (ER) and mitochondria. At these contact sites, EGFR-dependent, ER-generated Ca2+ oscillations are sensed by mitochondria, leading to increased metabolism and ATP production. Locally released ATP is required for cortical actin remodeling and EGFR-NCE vesicle fission. The same biochemical circuitry is also needed for an effector function of EGFR, i.e., collective motility. The multiorganelle signaling platform herein described mediates direct communication between EGFR signaling and mitochondrial metabolism, and is predicted to have a broad impact on cell physiology as it is activated by another growth factor receptor, HGFR/MET.
Collapse
Affiliation(s)
- Deborah Mesa
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Andrea Raimondi
- Experimental Imaging Centre, IRCCS San Raffaele Hospital Scientific Institute, Milan, Italy
- Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Stefano Freddi
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Gorana Jendrisek
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Micaela Quarto
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Irene Schiano Lomoriello
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Grazia Malabarba
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Arianna Bresci
- Department of Physics, Politecnico di Milano, Milan, Italy
| | | | | | - Hind Abdo
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Giorgio Scita
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Candiolo, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Dario Polli
- Department of Physics, Politecnico di Milano, Milan, Italy
- CNR Institute for Photonics and Nanotechnology (CNR-IFN), Milan, Italy
| | - Carlo Tacchetti
- Experimental Imaging Centre, IRCCS San Raffaele Hospital Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Section of Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Pier Paolo Di Fiore
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| | - Sara Sigismund
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy.
- IEO, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
21
|
Dai M, Chen Y, Qin J. Atrioventricular re-entrant tachycardia and atrioventricular node re-entrant tachycardia in a patient with cancer under chemotherapy: a case report and literature review. Front Cardiovasc Med 2024; 11:1367893. [PMID: 38911514 PMCID: PMC11190324 DOI: 10.3389/fcvm.2024.1367893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
Cardio-oncology is a new field of interest in cardiology focusing on the detection and treatment of cardiovascular diseases, such as arrhythmias, myocarditis, and heart failure, as side-effects of chemotherapy and radiotherapy. The association between chemotherapeutic agents and arrhythmias has previously been established. Atrial tachyarrhythmias, particularly atrial fibrillation, are most common, but ventricular arrhythmias, including those related to treatment-induced QT prolongation, and bradyarrhythmias can also occur. However, the association between chemotherapeutic agents and atrioventricular re-entrant tachycardia (AVRT)/atrioventricular node re-entrant tachycardia (AVNRT) remains poorly understood. Here, we report a patient with new-onset AVRT/AVNRT and lung cancer who underwent chemotherapy. We considered that chemotherapy or cancer itself may have been a trigger for the initiation of paroxysmal AVRT/AVNRT, and that radiofrequency catheter ablation was effective in treating this type of tachycardia. Here, possible mechanisms and potential genes (mostly ion channels) involved in AVRT/AVNRT are summarized and the mechanisms underlying the possible regulatory patterns of cancer cells and chemotherapy on ion channels are reviewed. Finally, we considered that ion channel abnormalities may link cancer or chemotherapy to the onset of AVRT/AVNRT. The aim of the present study was to highlight the association between chemotherapeutic agents and AVRT/AVNRT and to provide new insights for future research. Understanding the intermediate mechanisms between chemotherapeutic agents and AVRT/AVNRT may be beneficial in preventing chemotherapy-evoked AVRT/AVNRT (and/or other arrhythmias) in future.
Collapse
Affiliation(s)
- Meiyan Dai
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chen
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin Qin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Lv X, Lan G, Guo Q. Identification of Subtypes in Triple-negative Breast Cancer Based on Shared Genes Between Immunity and Cancer Stemness. J Immunother 2024; 47:107-116. [PMID: 38369822 DOI: 10.1097/cji.0000000000000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/13/2023] [Indexed: 02/20/2024]
Abstract
The correlation between triple-negative breast cancer (TNBC) and genes related to immunity and cancer stemness, particularly shared genes, remains unclear. This study aimed to investigate the correlation of immunity and cancer stemness with the molecular subtyping and survival rates in TNBC using bioinformatics approaches. Differential gene analysis was conducted to identify TNBC-associated differentially expressed genes (DEGs). Cancer stem cell (CSC)-related genes were obtained using weighted gene coexpression network analysis. Immune-related gene sets were retrieved from the literature. Venn analysis was performed to identify the shared DEGs between immunity and cancer stemness in TNBC. Cluster analysis and survival analysis based on the expression of these genes were conducted to identify TNBC subtypes with significant survival differences. A total of 5259 TNBC-associated DEGs, 2214 CSC-related genes, 1793 immune-related genes, and 44 shared DEGs between immunity and cancer stemness were obtained. Among them, 3 shared DEGs were closely associated with TNBC survival rates ( P <0.05). Cluster and survival analyses revealed that among 3 subtypes, cluster2 exhibited the best survival rate, and cluster3 showed the worst survival rate ( P <0.05). Dendritic cells were highly infiltrated in cluster2, while plasma cells and resting mast cells were highly infiltrated in cluster3 ( P <0.05). Genes shared by immunity and cancer stemness were capable of classifying TNBC samples. TNBC patients of different subtypes exhibited significant differences in immune profiles, genetic mutations, and drug sensitivity. These findings could provide new insights into the pathogenesis of TNBC, the immune microenvironment, and the selection of therapeutic targets for drug treatment.
Collapse
Affiliation(s)
- Xianmei Lv
- Department of Radiotherapy, Jinhua People's Hospital, Jinhua, China
| | - Gaochen Lan
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiusheng Guo
- Department of Medical Oncology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
23
|
De Mario A, Trevellin E, Piazza I, Vindigni V, Foletto M, Rizzuto R, Vettor R, Mammucari C. Mitochondrial Ca 2+ signaling is a hallmark of specific adipose tissue-cancer crosstalk. Sci Rep 2024; 14:8469. [PMID: 38605098 PMCID: PMC11009327 DOI: 10.1038/s41598-024-55650-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 04/13/2024] Open
Abstract
Obesity is associated with increased risk and worse prognosis of many tumours including those of the breast and of the esophagus. Adipokines released from the peritumoural adipose tissue promote the metastatic potential of cancer cells, suggesting the existence of a crosstalk between the adipose tissue and the surrounding tumour. Mitochondrial Ca2+ signaling contributes to the progression of carcinoma of different origins. However, whether adipocyte-derived factors modulate mitochondrial Ca2+ signaling in tumours is unknown. Here, we show that conditioned media derived from adipose tissue cultures (ADCM) enriched in precursor cells impinge on mitochondrial Ca2+ homeostasis of target cells. Moreover, in modulating mitochondrial Ca2+ responses, a univocal crosstalk exists between visceral adipose tissue-derived preadipocytes and esophageal cancer cells, and between subcutaneous adipose tissue-derived preadipocytes and triple-negative breast cancer cells. An unbiased metabolomic analysis of ADCM identified creatine and creatinine for their ability to modulate mitochondrial Ca2+ uptake, migration and proliferation of esophageal and breast tumour cells, respectively.
Collapse
Affiliation(s)
- Agnese De Mario
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, 35131, Padua, Italy
| | - Elisabetta Trevellin
- Internal Medicine Unit, Department of Medicine, Padua University Hospital, via Giustiniani 2, 35128, Padua, Italy
| | - Ilaria Piazza
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, 35131, Padua, Italy
| | - Vincenzo Vindigni
- Clinic of Plastic and Reconstructive Surgery, Department of Neurosciences, University of Padua, Padua, Italy
| | - Mirto Foletto
- Bariatric Unit, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, 35131, Padua, Italy
| | - Roberto Vettor
- Internal Medicine Unit, Department of Medicine, Padua University Hospital, via Giustiniani 2, 35128, Padua, Italy.
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, 35131, Padua, Italy.
| |
Collapse
|
24
|
Mitaishvili E, Feinsod H, David Z, Shpigel J, Fernandez C, Sauane M, de la Parra C. The Molecular Mechanisms behind Advanced Breast Cancer Metabolism: Warburg Effect, OXPHOS, and Calcium. FRONT BIOSCI-LANDMRK 2024; 29:99. [PMID: 38538285 PMCID: PMC10999756 DOI: 10.31083/j.fbl2903099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/24/2024] [Accepted: 02/22/2024] [Indexed: 04/10/2024]
Abstract
Altered metabolism represents a fundamental difference between cancer cells and normal cells. Cancer cells have a unique ability to reprogram their metabolism by deviating their reliance from primarily oxidative phosphorylation (OXPHOS) to glycolysis, in order to support their survival. This metabolic phenotype is referred to as the "Warburg effect" and is associated with an increase in glucose uptake, and a diversion of glycolytic intermediates to alternative pathways that support anabolic processes. These processes include synthesis of nucleic acids, lipids, and proteins, necessary for the rapidly dividing cancer cells, sustaining their growth, proliferation, and capacity for successful metastasis. Triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer, with the poorest patient outcome due to its high rate of metastasis. TNBC is characterized by elevated glycolysis and in certain instances, low OXPHOS. This metabolic dysregulation is linked to chemotherapeutic resistance in TNBC research models and patient samples. There is more than a single mechanism by which this metabolic switch occurs and here, we review the current knowledge of relevant molecular mechanisms involved in advanced breast cancer metabolism, focusing on TNBC. These mechanisms include the Warburg effect, glycolytic adaptations, microRNA regulation, mitochondrial involvement, mitochondrial calcium signaling, and a more recent player in metabolic regulation, JAK/STAT signaling. In addition, we explore some of the drugs and compounds targeting cancer metabolic reprogramming. Research on these mechanisms is highly promising and could ultimately offer new opportunities for the development of innovative therapies to treat advanced breast cancer characterized by dysregulated metabolism.
Collapse
Affiliation(s)
- Erna Mitaishvili
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
| | - Hanna Feinsod
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Zachary David
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Jessica Shpigel
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Chelsea Fernandez
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Moira Sauane
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
- Department of Biological Sciences, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
| | - Columba de la Parra
- Department of Chemistry, Herbert H. Lehman College, City University of New York, New York, NY 10468, USA
- PhD Program in Biology, The Graduate Center, City University of New York, New York, NY 10016, USA
- PhD Programs in Biochemistry and Chemistry, The Graduate Center, City University of New York, New York, NY 10016, USA
| |
Collapse
|
25
|
Marinas IC, Ignat L, Maurușa IE, Gaboreanu MD, Adina C, Popa M, Chifiriuc MC, Angheloiu M, Georgescu M, Iacobescu A, Pircalabioru GG, Stan M, Pinteala M. Insights into the physico-chemical and biological characterization of sodium lignosulfonate - silver nanosystems designed for wound management. Heliyon 2024; 10:e26047. [PMID: 38384565 PMCID: PMC10878957 DOI: 10.1016/j.heliyon.2024.e26047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
Chronic wounds represent one of the complications that might occur from the disruption of wound healing process. Recently, there has been a rise in interest in employing nanotechnology to develop novel strategies for accelerating wound healing. The aim of the present study was to use a green synthesis method to obtain AgNPs/NaLS systems useful for wounds management and perform an in-depth investigation of their behavior during and post-synthesis as well as of their biological properties. The colloids obtained from silver nanoparticles (AgNPs) and commercial sodium lignosulfonate (NaLS) in a single-pot aqueous procedure have been fully characterized by UV-Vis, FT-IR, DLS, TEM, XRD, and XPS to evaluate the synthesis efficiency and to provide new insights in the process of AgNPs formation and NaLS behavior in aqueous solutions. The effects of various concentrations of NaLS (0-16 mg/mL) and AgNO3 (0-20 mM) and of two different temperatures on AgNPs formation have been analyzed. Although the room temperature is feasible for AgNPs synthesis, the short mixing at 70 °C significantly increases the speed of nanoparticle formation and storage stability. In all experimental conditions AgNPs of 20-40 nm in size have been obtained. The antimicrobial activity assessed quantitatively on clinical and reference bacterial strains, both in suspension and biofilm growth state, revealed a broad antimicrobial spectrum, the most intensive inhibitory effect being noticed against Pseudomonas aeruginosa and Escherichia coli strains. The AgNP/NaLS enhanced the NO extracellular release, potentially contributing to the microbicidal and anti-adherence activity by protein oxidation. Both AgNP/NaLS and NaLS were non-hemolytic (hemolytic index<5%, 2.26 ± 0.13% hemolysis) and biocompatible (102.17 ± 3.43 % HaCaT cells viability). The presence of AgNPs increased the antioxidative activity and induced a significant cytotoxicity on non-melanoma skin cancer cells (62.86 ± 8.27% Cal-27 cells viability). Taken together, all these features suggest the multivalent potential of these colloids for the development of novel strategies for wound management, acting by preventing infection-associated complications and supporting the tissue regeneration.
Collapse
Affiliation(s)
- Ioana C. Marinas
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Research and Development Department of SC Sanimed International Impex SRL, 6 Bucharest -Giurgiu Street, 087040, Giurgiu, Romania
| | - Leonard Ignat
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, Gr. Ghica Voda Alley 41A, Iasi, 700487, Romania
| | - Ignat E. Maurușa
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, Gr. Ghica Voda Alley 41A, Iasi, 700487, Romania
| | - Madalina D. Gaboreanu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Faculty of Biology, Department of Botany and Microbiology, University of Bucharest, 1-3 Portocalelor Street, 060101, Bucharest, Romania
| | - Coroabă Adina
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, Gr. Ghica Voda Alley 41A, Iasi, 700487, Romania
| | - Marcela Popa
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Faculty of Biology, Department of Botany and Microbiology, University of Bucharest, 1-3 Portocalelor Street, 060101, Bucharest, Romania
| | - Mariana C. Chifiriuc
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Faculty of Biology, Department of Botany and Microbiology, University of Bucharest, 1-3 Portocalelor Street, 060101, Bucharest, Romania
- Romanian Academy of Scientists, 54 Spl. Independentei St., District 5, 50085, Bucharest, Romania
- The Romanian Academy, 25, Calea Victoriei, Sector 1, District 1, 010071, Bucharest, Romania
| | - Marian Angheloiu
- Research and Development Department of SC Sanimed International Impex SRL, 6 Bucharest -Giurgiu Street, 087040, Giurgiu, Romania
| | - Mihaela Georgescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
| | - Alexandra Iacobescu
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, Gr. Ghica Voda Alley 41A, Iasi, 700487, Romania
| | - Gratiela Gradisteanu Pircalabioru
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Faculty of Biology, Department of Botany and Microbiology, University of Bucharest, 1-3 Portocalelor Street, 060101, Bucharest, Romania
| | - Miruna Stan
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050095, Bucharest, Romania
- Faculty of Biology, Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095, Bucharest, Romania
| | - Mariana Pinteala
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, “Petru Poni” Institute of Macromolecular Chemistry, Gr. Ghica Voda Alley 41A, Iasi, 700487, Romania
| |
Collapse
|
26
|
Shamis SA, Savioli F, Ammar A, Al-Badran SS, Hatthakarnkul P, Leslie H, Mallon EE, Jamieson NB, McMillan DC, Edwards J. Spatial transcriptomic analysis of tumour with high and low CAIX expression in TNBC tissue samples using GeoMx™ RNA assay. Histol Histopathol 2024; 39:177-200. [PMID: 37681672 DOI: 10.14670/hh-18-655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
PURPOSE Prognostic significance and gene signatures associated with carbonic anhydrase IX (CAIX) was investigated in triple negative breast cancer (TNBC) patients. METHODS Immunohistochemistry (IHC) for CAIX was performed in tissue microarrays (TMAs) of 136 TNBC patients. In a subset of 52 patients Digital Spatial Profiler (DSP) was performed in tumour (pan-cytokeratin+) and stroma (pan-cytokeratin-). Differentially expressed genes (DEGs) with P<0.05 and and log2 fold change (FC)>(±0.25 and ±0.3, for tumour and stromal compartment, respectively) were identified. Four genes were validated at the protein level. RESULT Cytoplasmic CAIX expression was independently associated with poor recurrence free survival in TNBC patients [hazard ratio (HR)=6.59, 95% confidence interval (CI): 1.47-29.58, P=0.014]. DEG analysis identified 4 up-regulated genes (CD68, HIF1A, pan-melanocyte, and VSIR) in the tumour region and 9 down-regulated genes in the stromal region (CD86, CD3E, MS4A1, BCL2, CCL5, NKG7, PTPRC, CD27, and FAS) when low versus high CAIX expression was explored. Employing IHC, high CD68 and HIF-1α was associated with poorer prognosis and high BCL2 and CD3 was associated with good prognosis. CONCLUSIONS DSP technology identified DEGs in TNBC. Selected genes validated by IHC showed involvement of CD3 and BCL2 expression within stroma and HIF-1α, and CD68 expression within tumour. However, further functional analysis is warranted.
Collapse
Affiliation(s)
- Suad Ak Shamis
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Alexandria Parade, Glasgow, United Kingdom.
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Francesca Savioli
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Alexandria Parade, Glasgow, United Kingdom.
| | - Aula Ammar
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Sara Sf Al-Badran
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Phimmada Hatthakarnkul
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Holly Leslie
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Elizabeth Ea Mallon
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Nigel B Jamieson
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Alexandria Parade, Glasgow, United Kingdom.
| | - Joanne Edwards
- Unit of Molecular Pathology, School of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Glasgow, United Kingdom
| |
Collapse
|
27
|
An G, Park J, Song J, Hong T, Song G, Lim W. Relevance of the endoplasmic reticulum-mitochondria axis in cancer diagnosis and therapy. Exp Mol Med 2024; 56:40-50. [PMID: 38172597 PMCID: PMC10834980 DOI: 10.1038/s12276-023-01137-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2024] Open
Abstract
Dynamic interactions between organelles are responsible for a variety of intercellular functions, and the endoplasmic reticulum (ER)-mitochondrial axis is recognized as a representative interorganelle system. Several studies have confirmed that most proteins in the physically tethered sites between the ER and mitochondria, called mitochondria-associated ER membranes (MAMs), are vital for intracellular physiology. MAM proteins are involved in the regulation of calcium homeostasis, lipid metabolism, and mitochondrial dynamics and are associated with processes related to intracellular stress conditions, such as oxidative stress and unfolded protein responses. Accumulating evidence has shown that, owing to their extensive involvement in cellular homeostasis, alterations in the ER-mitochondrial axis are one of the etiological factors of tumors. An in-depth understanding of MAM proteins and their impact on cell physiology, particularly in cancers, may help elucidate their potential as diagnostic and therapeutic targets for cancers. For example, the modulation of MAM proteins is utilized not only to target diverse intracellular signaling pathways within cancer cells but also to increase the sensitivity of cancer cells to anticancer reagents and regulate immune cell activities. Therefore, the current review summarizes and discusses recent advances in research on the functional roles of MAM proteins and their characteristics in cancers from a diagnostic perspective. Additionally, this review provides insights into diverse therapeutic strategies that target MAM proteins in various cancer types.
Collapse
Affiliation(s)
- Garam An
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Junho Park
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
28
|
Konjalwar S, Ceyhan B, Rivera O, Nategh P, Neghabi M, Pavlovic M, Allani S, Ranji M. Demonstrating drug treatment efficacies by monitoring superoxide dynamics in human lung cancer cells with time-lapse fluorescence microscopy. JOURNAL OF BIOPHOTONICS 2024; 17:e202300331. [PMID: 37822188 PMCID: PMC12013861 DOI: 10.1002/jbio.202300331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
Metformin hydrochloride, an antihyperglycemic agent, and sulindac, a nonsteroidal anti-inflammatory drug, are FDA-approved drugs known to exert anticancer effects. Previous studies demonstrated sulindac and metformin's anticancer properties through mitochondrial dysfunction and inhibition of mitochondrial electron transport chain complex I and key signaling pathways. In this study, various drugs were administered to A549 lung cancer cells, and results revealed that a combination of sulindac and metformin enhanced cell death compared to the administration of the drugs separately. To measure superoxide production over time, we employed a time-lapse fluorescence imaging technique using mitochondrial-targeted hydroethidine. Fluorescence microscopy data showed the most significant increases in superoxide production in the combination treatment of metformin and sulindac. Results showed significant differences between the combined drug treatment and control groups and between the positive control and control groups. This approach can be utilized to quantify the anticancer efficacy of drugs, creating possibilities for additional therapeutic options.
Collapse
Affiliation(s)
- Shalaka Konjalwar
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| | - Busenur Ceyhan
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| | - Oscar Rivera
- Center for Molecular Biology and Biotechnology at Florida Atlantic University, Boca Raton, United States of America
| | - Parisa Nategh
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| | - Mehrnoosh Neghabi
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| | - Mirjana Pavlovic
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| | - Shailaja Allani
- Center for Molecular Biology and Biotechnology at Florida Atlantic University, Boca Raton, United States of America
| | - Mahsa Ranji
- Department of Electrical Engineering and Computer Science at Florida Atlantic University, Boca Raton, United States of America
| |
Collapse
|
29
|
MacColl Garfinkel A, Mnatsakanyan N, Patel JH, Wills AE, Shteyman A, Smith PJS, Alavian KN, Jonas EA, Khokha MK. Mitochondrial leak metabolism induces the Spemann-Mangold Organizer via Hif-1α in Xenopus. Dev Cell 2023; 58:2597-2613.e4. [PMID: 37673063 PMCID: PMC10840693 DOI: 10.1016/j.devcel.2023.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
An instructive role for metabolism in embryonic patterning is emerging, although a role for mitochondria is poorly defined. We demonstrate that mitochondrial oxidative metabolism establishes the embryonic patterning center, the Spemann-Mangold Organizer, via hypoxia-inducible factor 1α (Hif-1α) in Xenopus. Hypoxia or decoupling ATP production from oxygen consumption expands the Organizer by activating Hif-1α. In addition, oxygen consumption is 20% higher in the Organizer than in the ventral mesoderm, indicating an elevation in mitochondrial respiration. To reconcile increased mitochondrial respiration with activation of Hif-1α, we discovered that the "free" c-subunit ring of the F1Fo ATP synthase creates an inner mitochondrial membrane leak, which decouples ATP production from respiration at the Organizer, driving Hif-1α activation there. Overexpression of either the c-subunit or Hif-1α is sufficient to induce Organizer cell fates even when β-catenin is inhibited. We propose that mitochondrial leak metabolism could be a general mechanism for activating Hif-1α and Wnt signaling.
Collapse
Affiliation(s)
- Alexandra MacColl Garfinkel
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Nelli Mnatsakanyan
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jeet H Patel
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98195, USA; Program in Molecular and Cellular Biology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Andrea E Wills
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Amy Shteyman
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Peter J S Smith
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | | | - Elizabeth Ann Jonas
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA.
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
30
|
D’Angelo D, Rizzuto R. The Mitochondrial Calcium Uniporter (MCU): Molecular Identity and Role in Human Diseases. Biomolecules 2023; 13:1304. [PMID: 37759703 PMCID: PMC10526485 DOI: 10.3390/biom13091304] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Calcium (Ca2+) ions act as a second messenger, regulating several cell functions. Mitochondria are critical organelles for the regulation of intracellular Ca2+. Mitochondrial calcium (mtCa2+) uptake is ensured by the presence in the inner mitochondrial membrane (IMM) of the mitochondrial calcium uniporter (MCU) complex, a macromolecular structure composed of pore-forming and regulatory subunits. MtCa2+ uptake plays a crucial role in the regulation of oxidative metabolism and cell death. A lot of evidence demonstrates that the dysregulation of mtCa2+ homeostasis can have serious pathological outcomes. In this review, we briefly discuss the molecular structure and the function of the MCU complex and then we focus our attention on human diseases in which a dysfunction in mtCa2+ has been shown.
Collapse
Affiliation(s)
- Donato D’Angelo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy;
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy;
- National Center on Gene Therapy and RNA-Based Drugs, 35131 Padua, Italy
| |
Collapse
|
31
|
Wu H, Chen W, Chen Z, Li X, Wang M. Novel tumor therapy strategies targeting endoplasmic reticulum-mitochondria signal pathways. Ageing Res Rev 2023; 88:101951. [PMID: 37164161 DOI: 10.1016/j.arr.2023.101951] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/13/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Organelles form tight connections through membrane contact sites, thereby cooperating to regulate homeostasis and cell function. Among them, the contact between endoplasmic reticulum (ER), the main intracellular calcium storage organelles, and mitochondria has been recognized for decades, and its main roles in the ion and lipid transport, ROS signaling, membrane dynamic changes and cellular metabolism are basically determined. At present, many tumor chemotherapeutic drugs rely on ER-mitochondrial calcium signal to function, but the mechanism of targeting resident molecules at the mitochondria-associated endoplasmic reticulum membranes (MAM) to sensitize traditional chemotherapy and the new tumor therapeutic targets identified based on the signal pathways on the MAM have not been thoroughly discussed. In this review, we highlight the key roles of various signaling pathways at the ER-mitochondria contact site in tumorigenesis and focus on novel anticancer therapy strategies targeting potential targets at this contact site.
Collapse
Affiliation(s)
- Hongzheng Wu
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wanxin Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhenni Chen
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xianping Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Wang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
32
|
Miao Y, Wang X, Lai Y, Huang Y, Yin H, Meng X, Liu H, Hou R, Lin W, Zhang X, Zhang X, Chai BC, Zhang F, Guo L, Yang S. Targeting the mitochondrial calcium uniporter inhibits cancer progression and alleviates cisplatin resistance in esophageal squamous cell carcinoma. Int J Oncol 2023; 63:82. [PMID: 37264968 PMCID: PMC10552700 DOI: 10.3892/ijo.2023.5530] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 05/05/2023] [Indexed: 06/03/2023] Open
Abstract
Cisplatin is the standard chemotherapeutic drug used for the treatment of esophageal squamous cell carcinoma (ESCC). Acquired cisplatin resistance is the primary obstacle to prolonging patient survival time. Here, the therapeutic effects of mitochondrial calcium uniporter (MCU) inhibition on tumor growth and cisplatin resistance in ESCC were assessed. MCU was stably overexpressed or knocked down in three ESCC cell lines and three cisplatin‑resistant ESCC cell lines. Then, proliferation, migration, and mitochondrial membrane potential (MMP) were measured by colony formation, wound healing, Transwell, and JC‑1 staining assays. MCU, MICU2, MICU1, and PD‑L1 levels were detected through western blotting and immunofluorescence. ESCC and cisplatin‑resistant ESCC xenograft mouse models were established. After MCU knockdown, tumor volume was measured. The expression levels of proliferation markers (CyclinD1 and Ki‑67), MICU1/2, PD‑L1, epithelial-mesenchymal transition (EMT) markers (vimentin, β‑catenin, and E‑cadherin), and the angiogenesis marker CD34 were detected through western blotting, immunohistochemistry, or immunofluorescence. The results showed that MCU overexpression significantly promoted proliferation, migration, and MMP in ESCC cells and cisplatin‑resistant ESCC cells. However, proliferation, migration, and MMP were suppressed following MCU knockdown. In ESCC cells, MCU overexpression markedly increased MICU2, MICU1, and PD‑L1 levels, and the opposite results were observed when MCU was stably knocked down. Similarly, MCU inhibition decreased MICU2, MICU1, and PD‑L1 expression in cisplatin‑resistant ESCC cells. Moreover, MCU knockdown substantially decreased tumor growth, EMT, and angiogenesis in ESCC and cisplatin‑resistant ESCC xenograft mice. Collectively, targeting MCU may inhibit cancer progression and alleviate cisplatin resistance in ESCC.
Collapse
Affiliation(s)
- Yu Miao
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Xiaofei Wang
- Pathology Department, North China University of Science and Technology Affiliated Hospital, Tangshan, Hebie 063000
| | - Yafang Lai
- Department of Gastroenterology, Ordos Central Hospital, Ordos, Inner Mongolia Autonomous Region 017000
| | - Ying Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Hua Yin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Xiangkun Meng
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Hao Liu
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Ruirui Hou
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Wan Lin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Xiaoxu Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Xu Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Bei Cho Chai
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Feixiong Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| | - Le Guo
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Shaoqi Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004
| |
Collapse
|
33
|
Zheng Q, Wang Y, Zhao R, Han P, Zhao J, Li L, Zhou X, Li P, Mo Y, Pan X, Luo W, Zhou X. Inactivation of epithelial sodium ion channel molecules serves as effective diagnostic biomarkers in clear cell renal cell carcinoma. Genes Genomics 2023; 45:855-866. [PMID: 37133722 DOI: 10.1007/s13258-023-01376-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 03/08/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND Non-voltage-gated sodium channel, also known as the epithelial sodium channel (ENaC), formed by heteromeric complexes consisting of SCNN1A, SCNN1B, and SCNN1G, is responsible for maintaining sodium ion and body fluid homeostasis in epithelial cells. However, no systematic study of SCNN1 family members has been conducted in renal clear cell carcinoma (ccRCC) to date. OBJECTIVE To investigate the abnormal expression of SCNN1 family in ccRCC and its potential correlation with clinical parameters. METHODS The transcription and protein expression levels of SCNN1 family members in ccRCC were analyzed based on the TCGA database, and were confirmed by quantitative RT-PCR and immunohistochemical staining assays, respectively. The area under curve (AUC) was used to evaluate the diagnostic value of SCNN1 family members for ccRCC patients. RESULTS The mRNA and protein expression of SCNN1 family members was significantly downregulated in ccRCC compared with normal kidney tissues, which might be due to DNA hypermethylation in the promoter region. It is worth noting that the AUC of SCNN1A, SCNN1B, and SCNN1G were 0.965, 0.979, and 0.988 based on the TCGA database (p < 0.0001), respectively. The diagnostic value was even higher when combing these three members together (AUC = 0.997, p < 0.0001). Intriguingly, the mRNA level of SCNN1A was significantly lower in females compared with males, while SCNN1B and SCNN1G were increased with the progression of ccRCC and remarkably associated with a worse outcome for patients. CONCLUSION The aberrantly decrease of SCNN1 family members might serve as valuable biomarkers for the diagnosis of ccRCC.
Collapse
Affiliation(s)
- Qian Zheng
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Yifang Wang
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Ran Zhao
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Peipei Han
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Jun Zhao
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Limei Li
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Xiaohui Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Ping Li
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Yingxi Mo
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Xinli Pan
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Academy of Sciences, Nanning, China
| | - Wenqi Luo
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, China.
| | - Xiaoying Zhou
- Life Science Institute, Guangxi Medical University, Nanning, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China.
| |
Collapse
|
34
|
Fernandez Garcia E, Paudel U, Noji MC, Bowman CE, Rustgi AK, Pitarresi JR, Wellen KE, Arany Z, Weissenrieder JS, Foskett JK. The mitochondrial Ca 2+ channel MCU is critical for tumor growth by supporting cell cycle progression and proliferation. Front Cell Dev Biol 2023; 11:1082213. [PMID: 37363724 PMCID: PMC10285664 DOI: 10.3389/fcell.2023.1082213] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Introduction: The mitochondrial uniporter (MCU) Ca2+ ion channel represents the primary means for Ca2+ uptake by mitochondria. Mitochondrial matrix Ca2+ plays critical roles in mitochondrial bioenergetics by impinging upon respiration, energy production and flux of biochemical intermediates through the TCA cycle. Inhibition of MCU in oncogenic cell lines results in an energetic crisis and reduced cell proliferation unless media is supplemented with nucleosides, pyruvate or α-KG. Nevertheless, the roles of MCU-mediated Ca2+ influx in cancer cells remain unclear, in part because of a lack of genetic models. Methods: MCU was genetically deleted in transformed murine fibroblasts for study in vitro and in vivo. Tumor formation and growth were studied in murine xenograft models. Proliferation, cell invasion, spheroid formation and cell cycle progression were measured in vitro. The effects of MCU deletion on survival and cell-death were determined by probing for live/death markers. Mitochondrial bioenergetics were studied by measuring mitochondrial matrix Ca2+ concentration, membrane potential, global dehydrogenase activity, respiration, ROS production and inactivating-phosphorylation of pyruvate dehydrogenase. The effects of MCU rescue on metabolism were examined by tracing of glucose and glutamine utilization for fueling of mitochondrial respiration. Results: Transformation of primary fibroblasts in vitro was associated with increased MCU expression, enhanced MCU-mediated Ca2+ uptake, altered mitochondrial matrix Ca2+ concentration responses to agonist stimulation, suppression of inactivating-phosphorylation of pyruvate dehydrogenase and a modest increase of mitochondrial respiration. Genetic MCU deletion inhibited growth of HEK293T cells and transformed fibroblasts in mouse xenograft models, associated with reduced proliferation and delayed cell-cycle progression. MCU deletion inhibited cancer stem cell-like spheroid formation and cell invasion in vitro, both predictors of metastatic potential. Surprisingly, mitochondrial matrix [Ca2+], membrane potential, global dehydrogenase activity, respiration and ROS production were unaffected. In contrast, MCU deletion elevated glycolysis and glutaminolysis, strongly sensitized cell proliferation to glucose and glutamine limitation, and altered agonist-induced cytoplasmic Ca2+ signals. Conclusion: Our results reveal a dependence of tumorigenesis on MCU, mediated by a reliance on MCU for cell metabolism and Ca2+ dynamics necessary for cell-cycle progression and cell proliferation.
Collapse
Affiliation(s)
- Emily Fernandez Garcia
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Usha Paudel
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael C. Noji
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Medicine, Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Caitlyn E. Bowman
- Department of Medicine, Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, United States
| | - Jason R. Pitarresi
- Division of Hematology/Oncology, Departments of Medicine and Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Kathryn E. Wellen
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zolt Arany
- Department of Medicine, Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Jillian S. Weissenrieder
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - J. Kevin Foskett
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
35
|
Bantsimba-Malanda C, Ahidouch A, Rodat-Despoix L, Ouadid-Ahidouch H. Calcium signal modulation in breast cancer aggressiveness. Cell Calcium 2023; 113:102760. [PMID: 37247443 DOI: 10.1016/j.ceca.2023.102760] [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: 03/22/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023]
Abstract
Breast cancer (BC) is the second most common cancer and cause of death in women. The aggressive subtypes including triple negative types (TNBCs) show a resistance to chemotherapy, impaired immune system, and a worse prognosis. From a histological point of view, TNBCs are deficient in oestrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2+) expression. Many studies reported an alteration in the expression of calcium channels, calcium binding proteins and pumps in BC that promote proliferation, survival, resistance to chemotherapy, and metastasis. Moreover, Ca2+ signal remodeling and calcium transporters expression have been associated to TNBCs and HER2+ BC subtypes. This review provides insight into the underlying alteration of the expression of calcium-permeable channels, pumps, and calcium dependent proteins and how this alteration plays an important role in promoting metastasis, metabolic switching, inflammation, and escape to chemotherapy treatment and immune surveillance in aggressive BC including TNBCs models and highly metastatic BC tumors.
Collapse
Affiliation(s)
- Claudie Bantsimba-Malanda
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne Amiens, France
| | - Ahmed Ahidouch
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne Amiens, France; Department of Biology, Faculty of Sciences, University Ibn Zohr, Agadir 80000, Morocco
| | - Lise Rodat-Despoix
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne Amiens, France.
| | - Halima Ouadid-Ahidouch
- Laboratory of Cellular and Molecular Physiology, UR UPJV 4667, University of Picardie Jules Verne Amiens, France.
| |
Collapse
|
36
|
García EF, Paudel U, Noji MC, Bowman CE, Pitarresi JR, Rustgi AK, Wellen KE, Arany Z, Weissenrieder JS, Foskett JK. The mitochondrial Ca 2+ channel MCU is critical for tumor growth by supporting cell cycle progression and proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.26.538295. [PMID: 37163088 PMCID: PMC10168388 DOI: 10.1101/2023.04.26.538295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The mitochondrial uniporter (MCU) Ca 2+ ion channel represents the primary means for Ca 2+ uptake into mitochondria. Here we employed in vitro and in vivo models with MCU genetically eliminated to understand how MCU contributes to tumor formation and progression. Transformation of primary fibroblasts in vitro was associated with increased MCU expression, enhanced mitochondrial Ca 2+ uptake, suppression of inactivating-phosphorylation of pyruvate dehydrogenase, a modest increase of basal mitochondrial respiration and a significant increase of acute Ca 2+ -dependent stimulation of mitochondrial respiration. Inhibition of mitochondrial Ca 2+ uptake by genetic deletion of MCU markedly inhibited growth of HEK293T cells and of transformed fibroblasts in mouse xenograft models. Reduced tumor growth was primarily a result of substantially reduced proliferation and fewer mitotic cells in vivo , and slower cell proliferation in vitro associated with delayed progression through S-phase of the cell cycle. MCU deletion inhibited cancer stem cell-like spheroid formation and cell invasion in vitro , both predictors of metastatic potential. Surprisingly, mitochondrial matrix Ca 2+ concentration, membrane potential, global dehydrogenase activity, respiration and ROS production were unchanged by genetic deletion of MCU in transformed cells. In contrast, MCU deletion elevated glycolysis and glutaminolysis, strongly sensitized cell proliferation to glucose and glutamine limitation, and altered agonist-induced cytoplasmic Ca 2+ signals. Our results reveal a dependence of tumorigenesis on MCU, mediated by a reliance on mitochondrial Ca 2+ uptake for cell metabolism and Ca 2+ dynamics necessary for cell-cycle progression and cell proliferation.
Collapse
|
37
|
Blacker TS, Duchen MR, Bain AJ. NAD(P)H binding configurations revealed by time-resolved fluorescence and two-photon absorption. Biophys J 2023; 122:1240-1253. [PMID: 36793214 PMCID: PMC10111271 DOI: 10.1016/j.bpj.2023.02.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/07/2022] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
NADH and NADPH play key roles in the regulation of metabolism. Their endogenous fluorescence is sensitive to enzyme binding, allowing changes in cellular metabolic state to be determined using fluorescence lifetime imaging microscopy (FLIM). However, to fully uncover the underlying biochemistry, the relationships between their fluorescence and binding dynamics require greater understanding. Here we accomplish this through time- and polarization-resolved fluorescence and polarized two-photon absorption measurements. Two lifetimes result from binding of both NADH to lactate dehydrogenase and NADPH to isocitrate dehydrogenase. The composite fluorescence anisotropy indicates the shorter (1.3-1.6 ns) decay component to be accompanied by local motion of the nicotinamide ring, pointing to attachment solely via the adenine moiety. For the longer lifetime (3.2-4.4 ns), the nicotinamide conformational freedom is found to be fully restricted. As full and partial nicotinamide binding are recognized steps in dehydrogenase catalysis, our results unify photophysical, structural, and functional aspects of NADH and NADPH binding and clarify the biochemical processes that underlie their contrasting intracellular lifetimes.
Collapse
Affiliation(s)
- Thomas S Blacker
- Department of Physics & Astronomy, University College London, London, United Kingdom; Research Department of Cell & Developmental Biology, University College London, London, United Kingdom
| | - Michael R Duchen
- Research Department of Cell & Developmental Biology, University College London, London, United Kingdom
| | - Angus J Bain
- Department of Physics & Astronomy, University College London, London, United Kingdom.
| |
Collapse
|
38
|
Filadi R, De Mario A, Audano M, Romani P, Pedretti S, Cardenas C, Dupont S, Mammucari C, Mitro N, Pizzo P. Sustained IP3-linked Ca2+ signaling promotes progression of triple negative breast cancer cells by regulating fatty acid metabolism. Front Cell Dev Biol 2023; 11:1071037. [PMID: 36994106 PMCID: PMC10040683 DOI: 10.3389/fcell.2023.1071037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Rewiring of mitochondrial metabolism has been described in different cancers as a key step for their progression. Calcium (Ca2+) signaling regulates mitochondrial function and is known to be altered in several malignancies, including triple negative breast cancer (TNBC). However, whether and how the alterations in Ca2+ signaling contribute to metabolic changes in TNBC has not been elucidated. Here, we found that TNBC cells display frequent, spontaneous inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ oscillations, which are sensed by mitochondria. By combining genetic, pharmacologic and metabolomics approaches, we associated this pathway with the regulation of fatty acid (FA) metabolism. Moreover, we demonstrated that these signaling routes promote TNBC cell migration in vitro, suggesting they might be explored to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Riccardo Filadi
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- *Correspondence: Riccardo Filadi, ,
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Patrizia Romani
- Department of Molecular Medicine (DMM), University of Padova, Padua, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Cesar Cardenas
- Faculty of Sciences, Universidad Mayor, Center for Integrative Biology, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, United States
| | - Sirio Dupont
- Department of Molecular Medicine (DMM), University of Padova, Padua, Italy
| | - Cristina Mammucari
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Myology Center (CIR-Myo), University of Padova, Padua, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Paola Pizzo
- Neuroscience Institute, National Research Council (CNR), Padua, Italy
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| |
Collapse
|
39
|
Pan D, Xu L, Shi D, Guo M. Knowledge mapping of mitochondrial calcium uniporter from 2011 to 2022: A bibliometric analysis. Front Physiol 2023; 14:1107328. [PMID: 36744031 PMCID: PMC9895098 DOI: 10.3389/fphys.2023.1107328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/11/2023] [Indexed: 01/21/2023] Open
Abstract
Background: Calcium uptake research has a long history. However, the mitochondrial calcium uniporter (MCU) protein was first discovered in 2011. As investigations of mitochondrial calcium uniporter represent a new research hotspot, a comprehensive and objective perspective of the field is lacking. Hence, this bibliometric analysis aimed to provide the current study status and trends related to mitochondrial calcium uniporter research in the past decade. Methods: Articles were acquired from the Web of Science Core Collection database. We quantified and visualized information regarding annual publications, journals, cocited journals, countries/regions, institutions, authors, and cocited authors by using CiteSpace 5.8. R3 and VOSviewer. In addition, we analysed the citation and keyword bursts related to mitochondrial calcium uniporter studies. Results: From 2011 to 2022, 1,030 articles were published by 5,050 authors from 1,145 affiliations and 62 countries or regions. The country with the most published articles was the United States. The institution with the most published articles was the University of Padua. Rosario Rizzuto published the most articles and was also the most cocited author. Cell Calcium published the largest number of articles, whereas Journal of Biological Chemistry had the most cocitations. The top 5 keywords related to pathological processes were oxidative stress, cell death, permeability transition, apoptosis, and metabolism. MICU1, calcium, ryanodine receptor, ATP synthase and cyclophilin D were the top 5 keywords related to molecules. Conclusion: mitochondrial calcium uniporter research has grown stably over the last decade. Current studies focus on the structure of the mitochondrial calcium uniporter complex and its regulatory effect on mitochondrial calcium homeostasis. In addition, the potential role of mitochondrial calcium uniporter in different diseases has been explored. Current studies mostly involve investigations of cancer and neurodegenerative diseases. Our analysis provides guidance and new insights into further mitochondrial calcium uniporter research.
Collapse
Affiliation(s)
- Deng Pan
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,Graduate School of Beijing University of Chinese Medicine, Beijing, China
| | - Lin Xu
- Gynecological Department of Traditional Chinese Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Dazhuo Shi, ; Ming Guo,
| | - Ming Guo
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China,*Correspondence: Dazhuo Shi, ; Ming Guo,
| |
Collapse
|
40
|
A Hypoxia Molecular Signature-Based Prognostic Model for Endometrial Cancer Patients. Int J Mol Sci 2023; 24:ijms24021675. [PMID: 36675190 PMCID: PMC9866886 DOI: 10.3390/ijms24021675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Endometrial cancer has the highest incidence of uterine corpus cancer, the sixth most typical cancer in women until 2020. High recurrence rate and frequent adverse events were reported in either standard chemotherapy or combined therapy. Hence, developing precise diagnostic and prognostic approaches for endometrial cancer was on demand. Four hypoxia-related genes were screened for the EC prognostic model by the univariate, LASSO, and multivariate Cox regression analysis from the TCGA dataset. QT-PCR and functional annotation analysis were performed. Associations between predicted risk and immunotherapy and chemotherapy responses were investigated by evaluating expressions of immune checkpoint inhibitors, infiltrated immune cells, m6a regulators, and drug sensitivity. The ROC curve and calibration plot indicated a fair predictability of our prognostic nomogram model. NR3C1 amplification, along with IL-6 and SRPX suppressions, were detected in tumor. High stromal score and enriched infiltrated aDCs and B cells in the high-risk group supported the hypothesis of immune-deserted tumor. Hypoxia-related molecular subtypes of EC were then identified via the gene signature. Cluster 2 patients showed a significant sensitivity to Vinblastine. In summary, our hypoxia signature model accurately predicted the survival outcome of EC patients and assessed translational and transcriptional dysregulations to explore targets for precise medical treatment.
Collapse
|
41
|
Colussi DM, Stathopulos PB. From passage to inhibition: Uncovering the structural and physiological inhibitory mechanisms of MCUb in mitochondrial calcium regulation. FASEB J 2023; 37:e22678. [PMID: 36538269 PMCID: PMC10107711 DOI: 10.1096/fj.202201080r] [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: 07/07/2022] [Revised: 10/14/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Mitochondrial calcium (Ca2+ ) regulation is critically implicated in the regulation of bioenergetics and cell fate. Ca2+ , a universal signaling ion, passively diffuses into the mitochondrial intermembrane space (IMS) through voltage-dependent anion channels (VDAC), where uptake into the matrix is tightly regulated across the inner mitochondrial membrane (IMM) by the mitochondrial Ca2+ uniporter complex (mtCU). In recent years, immense progress has been made in identifying and characterizing distinct structural and physiological mechanisms of mtCU component function. One of the main regulatory components of the Ca2+ selective mtCU channel is the mitochondrial Ca2+ uniporter dominant-negative beta subunit (MCUb). The structural mechanisms underlying the inhibitory effect(s) exerted by MCUb are poorly understood, despite high homology to the main mitochondrial Ca2+ uniporter (MCU) channel-forming subunits. In this review, we provide an overview of the structural differences between MCUb and MCU, believed to contribute to the inhibition of mitochondrial Ca2+ uptake. We highlight the possible structural rationale for the absent interaction between MCUb and the mitochondrial Ca2+ uptake 1 (MICU1) gatekeeping subunit and a potential widening of the pore upon integration of MCUb into the channel. We discuss physiological and pathophysiological information known about MCUb, underscoring implications in cardiac function and arrhythmia as a basis for future therapeutic discovery. Finally, we discuss potential post-translational modifications on MCUb as another layer of important regulation.
Collapse
Affiliation(s)
- Danielle M Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Peter B Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
42
|
Pharmacological targeting of the mitochondrial calcium-dependent potassium channel KCa3.1 triggers cell death and reduces tumor growth and metastasis in vivo. Cell Death Dis 2022; 13:1055. [PMID: 36539400 PMCID: PMC9768205 DOI: 10.1038/s41419-022-05463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Ion channels are non-conventional, druggable oncological targets. The intermediate-conductance calcium-dependent potassium channel (KCa3.1) is highly expressed in the plasma membrane and in the inner mitochondrial membrane (mitoKCa3.1) of various cancer cell lines. The role mitoKCa3.1 plays in cancer cells is still undefined. Here we report the synthesis and characterization of two mitochondria-targeted novel derivatives of a high-affinity KCa3.1 antagonist, TRAM-34, which retain the ability to block channel activity. The effects of these drugs were tested in melanoma, pancreatic ductal adenocarcinoma and breast cancer lines, as well as in vivo in two orthotopic models. We show that the mitochondria-targeted TRAM-34 derivatives induce release of mitochondrial reactive oxygen species, rapid depolarization of the mitochondrial membrane, fragmentation of the mitochondrial network. They trigger cancer cell death with an EC50 in the µM range, depending on channel expression. In contrast, inhibition of the plasma membrane KCa3.1 by membrane-impermeant Maurotoxin is without effect, indicating a specific role of mitoKCa3.1 in determining cell fate. At sub-lethal concentrations, pharmacological targeting of mitoKCa3.1 significantly reduced cancer cell migration by enhancing production of mitochondrial reactive oxygen species and nuclear factor-κB (NF-κB) activation, and by downregulating expression of Bcl-2 Nineteen kD-Interacting Protein (BNIP-3) and of Rho GTPase CDC-42. This signaling cascade finally leads to cytoskeletal reorganization and impaired migration. Overexpression of BNIP-3 or pharmacological modulation of NF-κB and CDC-42 prevented the migration-reducing effect of mitoTRAM-34. In orthotopic models of melanoma and pancreatic ductal adenocarcinoma, the tumors at sacrifice were 60% smaller in treated versus untreated animals. Metastasis of melanoma cells to lymph nodes was also drastically reduced. No signs of toxicity were observed. In summary, our results identify mitochondrial KCa3.1 as an unexpected player in cancer cell migration and show that its pharmacological targeting is efficient against both tumor growth and metastatic spread in vivo.
Collapse
|
43
|
Romito O, Guéguinou M, Raoul W, Champion O, Robert A, Trebak M, Goupille C, Potier-Cartereau M. Calcium signaling: A therapeutic target to overcome resistance to therapies in cancer. Cell Calcium 2022; 108:102673. [PMID: 36410063 DOI: 10.1016/j.ceca.2022.102673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Innate and acquired resistances to therapeutic agents are responsible for the failure of cancer treatments. Due to the multifactorial nature of resistance, the identification of new therapeutic targets is required to improve cancer treatment. Calcium is a universal second messenger that regulates many cellular functions such as proliferation, migration, and survival. Calcium channels, pumps and exchangers tightly regulate the duration, location and magnitude of calcium signals. Many studies have implicated dysregulation of calcium signaling in several pathologies, including cancer. Abnormal calcium fluxes due to altered channel expression or activation contribute to carcinogenesis and promote tumor development. However, there is limited information on the role of calcium signaling in cancer resistance to therapeutic drugs. This review discusses the role of calcium signaling as a mediator of cancer resistance, and assesses the potential value of combining anticancer therapy with calcium signaling modulators to improve the effectiveness of current treatments.
Collapse
Affiliation(s)
- Olivier Romito
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Maxime Guéguinou
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - William Raoul
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Ophélie Champion
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Alison Robert
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| | - Mohamed Trebak
- Vascular Medicine Institute, Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Caroline Goupille
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France; CHRU de Tours, hôpital Bretonneau, Tours, France.
| | - Marie Potier-Cartereau
- Inserm UMR 1069, Nutrition Croissance Cancer, Faculté de Médecine, Université de Tours, F-37032, France, Réseau 3MC « Molécules Marines, Métabolisme et Cancer » and Réseau CASTOR «Cancers des Tissus Hormono-Dépendants » Cancéropôle Grand Ouest, France.
| |
Collapse
|
44
|
Means RE, Katz SG. Balancing life and death: BCL-2 family members at diverse ER-mitochondrial contact sites. FEBS J 2022; 289:7075-7112. [PMID: 34668625 DOI: 10.1111/febs.16241] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 01/13/2023]
Abstract
The outer mitochondrial membrane is a busy place. One essential activity for cellular survival is the regulation of membrane integrity by the BCL-2 family of proteins. Another critical facet of the outer mitochondrial membrane is its close approximation with the endoplasmic reticulum. These mitochondrial-associated membranes (MAMs) occupy a significant fraction of the mitochondrial surface and serve as key signaling hubs for multiple cellular processes. Each of these pathways may be considered as forming their own specialized MAM subtype. Interestingly, like membrane permeabilization, most of these pathways play critical roles in regulating cellular survival and death. Recently, the pro-apoptotic BCL-2 family member BOK has been found within MAMs where it plays important roles in their structure and function. This has led to a greater appreciation that multiple BCL-2 family proteins, which are known to participate in numerous functions throughout the cell, also have roles within MAMs. In this review, we evaluate several MAM subsets, their role in cellular homeostasis, and the contribution of BCL-2 family members to their functions.
Collapse
Affiliation(s)
- Robert E Means
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Samuel G Katz
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
45
|
Stejerean‐Todoran I, Zimmermann K, Gibhardt CS, Vultur A, Ickes C, Shannan B, Bonilla del Rio Z, Wölling A, Cappello S, Sung H, Shumanska M, Zhang X, Nanadikar M, Latif MU, Wittek A, Lange F, Waters A, Brafford P, Wilting J, Urlaub H, Katschinski DM, Rehling P, Lenz C, Jakobs S, Ellenrieder V, Roesch A, Schön MP, Herlyn M, Stanisz H, Bogeski I. MCU
controls melanoma progression through a redox‐controlled phenotype switch. EMBO Rep 2022; 23:e54746. [PMID: 36156348 PMCID: PMC9638851 DOI: 10.15252/embr.202254746] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 01/16/2023] Open
Abstract
Melanoma is the deadliest of skin cancers and has a high tendency to metastasize to distant organs. Calcium and metabolic signals contribute to melanoma invasiveness; however, the underlying molecular details are elusive. The MCU complex is a major route for calcium into the mitochondrial matrix but whether MCU affects melanoma pathobiology was not understood. Here, we show that MCUA expression correlates with melanoma patient survival and is decreased in BRAF kinase inhibitor‐resistant melanomas. Knockdown (KD) of MCUA suppresses melanoma cell growth and stimulates migration and invasion. In melanoma xenografts, MCUA_KD reduces tumor volumes but promotes lung metastases. Proteomic analyses and protein microarrays identify pathways that link MCUA and melanoma cell phenotype and suggest a major role for redox regulation. Antioxidants enhance melanoma cell migration, while prooxidants diminish the MCUA_KD‐induced invasive phenotype. Furthermore, MCUA_KD increases melanoma cell resistance to immunotherapies and ferroptosis. Collectively, we demonstrate that MCUA controls melanoma aggressive behavior and therapeutic sensitivity. Manipulations of mitochondrial calcium and redox homeostasis, in combination with current therapies, should be considered in treating advanced melanoma.
Collapse
Affiliation(s)
- Ioana Stejerean‐Todoran
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | | | - Christine S Gibhardt
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Adina Vultur
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
- The Wistar Institute Melanoma Research Center Philadelphia PA USA
| | - Christian Ickes
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Batool Shannan
- The Wistar Institute Melanoma Research Center Philadelphia PA USA
- Department of Dermatology, University Hospital Essen, West German Cancer Center University Duisburg‐Essen and the German Cancer Consortium (DKTK)
| | - Zuriñe Bonilla del Rio
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Anna Wölling
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Sabrina Cappello
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Hsu‐Min Sung
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Magdalena Shumanska
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Xin Zhang
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Maithily Nanadikar
- Department of Cardiovascular Physiology, University Medical Center Göttingen Georg‐August‐University Göttingen Germany
| | - Muhammad U Latif
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology University Medical Center Göttingen Gottingen Germany
| | - Anna Wittek
- Department of NanoBiophotonics Max Planck Institute for Multidisciplinary Sciences Göttingen Germany
- Clinic of Neurology University Medical Center Göttingen Göttingen Germany
| | - Felix Lange
- Department of NanoBiophotonics Max Planck Institute for Multidisciplinary Sciences Göttingen Germany
- Clinic of Neurology University Medical Center Göttingen Göttingen Germany
| | - Andrea Waters
- The Wistar Institute Melanoma Research Center Philadelphia PA USA
| | | | - Jörg Wilting
- Department of Anatomy and Cell Biology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group Max Planck Institute for Multidisciplinary Sciences Göttingen Germany
- Bioanalytics, Institute of Clinical Chemistry University Medical Center Göttingen Germany
| | - Dörthe M Katschinski
- Department of Cardiovascular Physiology, University Medical Center Göttingen Georg‐August‐University Göttingen Germany
| | - Peter Rehling
- Department of Cellular Biochemistry University Medical Center Göttingen, GZMB Göttingen Germany
| | - Christof Lenz
- Bioanalytical Mass Spectrometry Group Max Planck Institute for Multidisciplinary Sciences Göttingen Germany
- Bioanalytics, Institute of Clinical Chemistry University Medical Center Göttingen Germany
| | - Stefan Jakobs
- Department of NanoBiophotonics Max Planck Institute for Multidisciplinary Sciences Göttingen Germany
- Clinic of Neurology University Medical Center Göttingen Göttingen Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology University Medical Center Göttingen Gottingen Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, West German Cancer Center University Duisburg‐Essen and the German Cancer Consortium (DKTK)
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Meenhard Herlyn
- The Wistar Institute Melanoma Research Center Philadelphia PA USA
| | - Hedwig Stanisz
- Department of Dermatology, Venereology and Allergology, University Medical Center Georg‐August‐University Göttingen Germany
| | - Ivan Bogeski
- Molecular Physiology, Department of Cardiovascular Physiology, University Medical Center Georg‐August‐University Göttingen Germany
| |
Collapse
|
46
|
Kuo CL, Ponneri Babuharisankar A, Lin YC, Lien HW, Lo YK, Chou HY, Tangeda V, Cheng LC, Cheng AN, Lee AYL. Mitochondrial oxidative stress in the tumor microenvironment and cancer immunoescape: foe or friend? J Biomed Sci 2022; 29:74. [PMID: 36154922 PMCID: PMC9511749 DOI: 10.1186/s12929-022-00859-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/19/2022] [Indexed: 12/07/2022] Open
Abstract
The major concept of "oxidative stress" is an excess elevated level of reactive oxygen species (ROS) which are generated from vigorous metabolism and consumption of oxygen. The precise harmonization of oxidative stresses between mitochondria and other organelles in the cell is absolutely vital to cell survival. Under oxidative stress, ROS produced from mitochondria and are the major mediator for tumorigenesis in different aspects, such as proliferation, migration/invasion, angiogenesis, inflammation, and immunoescape to allow cancer cells to adapt to the rigorous environment. Accordingly, the dynamic balance of oxidative stresses not only orchestrate complex cell signaling events in cancer cells but also affect other components in the tumor microenvironment (TME). Immune cells, such as M2 macrophages, dendritic cells, and T cells are the major components of the immunosuppressive TME from the ROS-induced inflammation. Based on this notion, numerous strategies to mitigate oxidative stresses in tumors have been tested for cancer prevention or therapies; however, these manipulations are devised from different sources and mechanisms without established effectiveness. Herein, we integrate current progress regarding the impact of mitochondrial ROS in the TME, not only in cancer cells but also in immune cells, and discuss the combination of emerging ROS-modulating strategies with immunotherapies to achieve antitumor effects.
Collapse
Affiliation(s)
- Cheng-Liang Kuo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Ananth Ponneri Babuharisankar
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Ying-Chen Lin
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Hui-Wen Lien
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Yu Kang Lo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Han-Yu Chou
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Vidhya Tangeda
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Li-Chun Cheng
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - An Ning Cheng
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan. .,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan. .,Department of Life Sciences, College of Health Sciences and Technology, National Central University, Zhongli, Taoyuan, 32001, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan. .,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
47
|
Rosa N, Speelman-Rooms F, Parys JB, Bultynck G. Modulation of Ca 2+ signaling by antiapoptotic Bcl-2 versus Bcl-xL: From molecular mechanisms to relevance for cancer cell survival. Biochim Biophys Acta Rev Cancer 2022; 1877:188791. [PMID: 36162541 DOI: 10.1016/j.bbcan.2022.188791] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/29/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Members of the Bcl-2-protein family are key controllers of apoptotic cell death. The family is divided into antiapoptotic (including Bcl-2 itself, Bcl-xL, Mcl-1, etc.) and proapoptotic members (Bax, Bak, Bim, Bim, Puma, Noxa, Bad, etc.). These proteins are well known for their canonical role in the mitochondria, where they control mitochondrial outer membrane permeabilization and subsequent apoptosis. However, several proteins are recognized as modulators of intracellular Ca2+ signals that originate from the endoplasmic reticulum (ER), the major intracellular Ca2+-storage organelle. More than 25 years ago, Bcl-2, the founding member of the family, was reported to control apoptosis through Ca2+ signaling. Further work elucidated that Bcl-2 directly targets and inhibits inositol 1,4,5-trisphosphate receptors (IP3Rs), thereby suppressing proapoptotic Ca2+ signaling. In addition to Bcl-2, Bcl-xL was also shown to impact cell survival by sensitizing IP3R function, thereby promoting prosurvival oscillatory Ca2+ release. However, new work challenges this model and demonstrates that Bcl-2 and Bcl-xL can both function as inhibitors of IP3Rs. This suggests that, depending on the cell context, Bcl-xL could support very distinct Ca2+ patterns. This not only raises several questions but also opens new possibilities for the treatment of Bcl-xL-dependent cancers. In this review, we will discuss the similarities and divergences between Bcl-2 and Bcl-xL regarding Ca2+ homeostasis and IP3R modulation from both a molecular and a functional point of view, with particular emphasis on cancer cell death resistance mechanisms.
Collapse
Affiliation(s)
- Nicolas Rosa
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular & Cellular Signaling, Department of Cellular & Molecular Medicine, Campus Gasthuisberg O/N-I bus 802, Herestraat 49, BE-3000 Leuven, Belgium.
| |
Collapse
|
48
|
Nair R, Gupta P, Shanmugam M. Mitochondrial metabolic determinants of multiple myeloma growth, survival, and therapy efficacy. Front Oncol 2022; 12:1000106. [PMID: 36185202 PMCID: PMC9523312 DOI: 10.3389/fonc.2022.1000106] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell dyscrasia characterized by the clonal proliferation of antibody producing plasma cells. Despite the use of next generation proteasome inhibitors (PI), immunomodulatory agents (IMiDs) and immunotherapy, the development of therapy refractory disease is common, with approximately 20% of MM patients succumbing to aggressive treatment-refractory disease within 2 years of diagnosis. A large emphasis is placed on understanding inter/intra-tumoral genetic, epigenetic and transcriptomic changes contributing to relapsed/refractory disease, however, the contribution of cellular metabolism and intrinsic/extrinsic metabolites to therapy sensitivity and resistance mechanisms is less well understood. Cancer cells depend on specific metabolites for bioenergetics, duplication of biomass and redox homeostasis for growth, proliferation, and survival. Cancer therapy, importantly, largely relies on targeting cellular growth, proliferation, and survival. Thus, understanding the metabolic changes intersecting with a drug's mechanism of action can inform us of methods to elicit deeper responses and prevent acquired resistance. Knowledge of the Warburg effect and elevated aerobic glycolysis in cancer cells, including MM, has allowed us to capitalize on this phenomenon for diagnostics and prognostics. The demonstration that mitochondria play critical roles in cancer development, progression, and therapy sensitivity despite the inherent preference of cancer cells to engage aerobic glycolysis has re-invigorated deeper inquiry into how mitochondrial metabolism regulates tumor biology and therapy efficacy. Mitochondria are the sole source for coupled respiration mediated ATP synthesis and a key source for the anabolic synthesis of amino acids and reducing equivalents. Beyond their core metabolic activities, mitochondria facilitate apoptotic cell death, impact the activation of the cytosolic integrated response to stress, and through nuclear and cytosolic retrograde crosstalk maintain cell fitness and survival. Here, we hope to shed light on key mitochondrial functions that shape MM development and therapy sensitivity.
Collapse
|
49
|
MCU Inhibitor Ruthenium Red Alleviates the Osteoclastogenesis and Ovariectomized Osteoporosis via Suppressing RANKL-Induced ROS Production and NFATc1 Activation through P38 MAPK Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7727006. [PMID: 36148414 PMCID: PMC9489416 DOI: 10.1155/2022/7727006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/05/2022] [Accepted: 08/18/2022] [Indexed: 12/04/2022]
Abstract
Osteoporosis is a disorder of bone metabolism that is extremely common in elderly patients as well as in postmenopausal women. The main manifestation is that the bone resorption capacity is greater than the bone formation capacity, which eventually leads to a decrease in bone mass, increasing the risk of fracture. There is growing evidence that inhibiting osteoclast formation and resorption ability can be effective in treating and preventing the occurrence of osteoporosis. Our study is the first time to explore the role of the mitochondrial calcium uniporter (MCU) and its inhibitor ruthenium red (RR) in bone metabolism, clarifying the specific mechanism by which it inhibits osteoclast formation in vitro and plays a therapeutic role in osteoporosis in vivo. We verified the suppressive effects of RR on the receptor activator of nuclear factor-κB ligand (RANKL-)-induced differentiation and bone resorption function of osteoclasts in vitro. The reactive oxygen species (ROS) production stimulated by RANKL and the expression level of P38 MAPK/NFATc1 were also found to be inhibited by RR. Moreover, the promotion of RR on osteogenesis differentiation was investigated by alkaline phosphatase (ALP) and alizarin red S (ARS) staining and the detection of osteogenesis-specific gene expression levels by quantitative polymerase chain reaction (qPCR) and western blotting. Moreover, in ovariectomy (OVX-)-induced osteoporosis models, RR can downregulate the expression and function of the MCU, relieving bone loss and promoting osteogenesis to present a therapeutic effect on osteoporosis. This new finding will provide an important direction for the study of RR and MCU in the study of bone metabolism therapy targets.
Collapse
|
50
|
Zhao J, Li M, Xu J, Cheng W. The modulation of ion channels in cancer chemo-resistance. Front Oncol 2022; 12:945896. [PMID: 36033489 PMCID: PMC9399684 DOI: 10.3389/fonc.2022.945896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Ion channels modulate the flow of ions into and out of a cell or intracellular organelle, leading to generation of electrical or chemical signals and regulating ion homeostasis. The abundance of ion channels in the plasma and intracellular membranes are subject to physiological and pathological regulations. Abnormal and dysregulated expressions of many ion channels are found to be linked to cancer and cancer chemo-resistance. Here, we will summarize ion channels distribution in multiple tumors. And the involvement of ion channels in cancer chemo-resistance will be highlighted.
Collapse
|