1
|
Špaková I, Smolko L, Sabolová G, Badovská Z, Kalinová K, Madreiter-Sokolowski C, Graier WF, Mareková M, Vašková J, Rabajdová M. Selective targeting of genes regulated by zinc finger proteins in endometriosis and endometrioid adenocarcinoma by zinc niflumato complex with neocuproine. Sci Rep 2025; 15:10126. [PMID: 40128272 PMCID: PMC11933352 DOI: 10.1038/s41598-025-94249-x] [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: 06/11/2024] [Accepted: 03/12/2025] [Indexed: 03/26/2025] Open
Abstract
Inadequate angiogenesis of endometriotic implants stimulated by the inflammatory microenvironment in the uterine region leads to the development of gynecological diseases, which significantly reduce the fertility and vitality of young women. Angiogenic processes are controlled by factors whose activities are regulated at the gene level by reactive oxygen species (ROS), hypoxia-induced factors (HIFs), and zinc-finger proteins (ZnFs) or posttranscriptionally via non-coding RNAs. The cooperation of these factors is responsible for the manifestation of pathological stimuli in the form of endometriosis of the body of the uterus, ovaries, or peritoneum, from which endometrioid carcinoma can develop. Molecules that can control gene expression by their intercalation to target DNA sequence, such as [Zn(neo)(nif)2], could prevent the hyperactivation of pro-angiogenic pathways (decrease HIF-1α, VEGF-A, TGF-β1, COX2, and ANG2/ANG1), reduce the formation of ROS, and reduce the risk of uterine neoplasticity. The NSAID-metal complex [Zn(neo)(nif)2] shows an ability to intercalate into ZNF3-7 target DNA sequence at a higher rate, which could explain its effect on genes regulated by this transcription factor. In addition, [Zn(neo)(nif)2] affects ROS production and Ca2+ level, possibly pointing to mitochondrial dysfunction as a potential cause for the described apoptosis.
Collapse
Affiliation(s)
- Ivana Špaková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Lukáš Smolko
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Gabriela Sabolová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Zuzana Badovská
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Katarína Kalinová
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Corina Madreiter-Sokolowski
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biohemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, T8010, Graz, Austria
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Janka Vašková
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, P. J. Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia.
| |
Collapse
|
2
|
Tawfik I, Schlick K, Ostaku J, Bresilla D, Gabrijelčič S, Gottschalk B, Sokolowski A, Malle E, Kalinova K, Hirtl M, Madreiter-Sokolowski CT. Breast cancer cells utilize T3 to trigger proliferation through cellular Ca 2+ modulation. Cell Commun Signal 2024; 22:533. [PMID: 39506805 PMCID: PMC11539297 DOI: 10.1186/s12964-024-01917-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024] Open
Abstract
High levels of thyroid hormones are linked to increased risk and advanced stages of breast cancer. Our previous work demonstrated that the biologically active triiodothyronine (T3) facilitates mitochondrial ATP production by upregulating Ca2+ handling proteins, thereby boosting mitochondrial Ca2+ uptake and Krebs cycle activity. In this study, different cell types were utilized to investigate whether T3 activates a Ca2+-induced signaling pathway to boost cancer cell proliferation. Using live-cell imaging, biochemical assays, and molecular profiling, differences in intracellular signaling among MCF7 and MDA-MB-468 breast cancer cells, non-cancerous breast cells hTERT-HME1, and PC3 prostate carcinoma cells, previously found to be insensitive to thyroid hormones in terms of proliferation, were investigated. Our findings revealed that T3 upregulates 1,4,5-trisphosphate receptor 3 via thyroid hormone receptor α. This boosts mitochondrial Ca2+ uptake, reduction equivalent yield, and mitochondrial ATP production, supporting the viability and proliferation of breast cancer cells without affecting non-cancerous hTERT-HME1 or PC3 prostate carcinoma cells. Understanding the interplay between T3 signaling, organellar interaction, and breast cancer metabolism could lead to targeted therapies that exploit cancer cell vulnerabilities. Our findings highlight T3 as a crucial regulator of cancer metabolism, reinforcing its potential as a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Ines Tawfik
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Katharina Schlick
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Julian Ostaku
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Doruntina Bresilla
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Sonja Gabrijelčič
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Benjamin Gottschalk
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Alwin Sokolowski
- Division of Restorative Dentistry, Periodontology and Prosthodontics, Medical University of Graz, Billrothgasse 4, Graz, 8010, Austria
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Katarina Kalinova
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Martin Hirtl
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria
| | - Corina T Madreiter-Sokolowski
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/IV, Graz, 8010, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
3
|
Butler LM, Evergren E. Ultrastructural analysis of prostate cancer tissue provides insights into androgen-dependent adaptations to membrane contact site establishment. Front Oncol 2023; 13:1217741. [PMID: 37529692 PMCID: PMC10389664 DOI: 10.3389/fonc.2023.1217741] [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: 05/05/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023] Open
Abstract
Membrane trafficking and organelle contact sites are important for regulating cell metabolism and survival; processes often deregulated in cancer. Prostate cancer is the second leading cause of cancer-related death in men in the developed world. While early-stage disease is curable by surgery or radiotherapy there is an unmet need to identify prognostic biomarkers, markers to treatment response and new therapeutic targets in intermediate-late stage disease. This study explored the morphology of organelles and membrane contact sites in tumor tissue from normal, low and intermediate histological grade groups. The morphology of organelles in secretory prostate epithelial cells; including Golgi apparatus, ER, lysosomes; was similar in prostate tissue samples across a range of Gleason scores. Mitochondrial morphology was not dramatically altered, but the number of membrane contacts with the ER notably increased with disease progression. A three-fold increase of tight mitochondria-ER membrane contact sites was observed in the intermediate Gleason score group compared to normal tissue. To investigate whether these changes were concurrent with an increased androgen signaling in the tissue, we investigated whether an anti-androgen used in the clinic to treat advanced prostate cancer (enzalutamide) could reverse the phenotype. Patient-derived explant tissues with an intermediate Gleason score were cultured ex vivo in the presence or absence of enzalutamide and the number of ER-mitochondria contacts were quantified for each matched pair of tissues. Enzalutamide treated tissue showed a significant reduction in the number and length of mitochondria-ER contact sites, suggesting a novel androgen-dependent regulation of these membrane contact sites. This study provides evidence for the first time that prostate epithelial cells undergo adaptations in membrane contact sites between mitochondria and the ER during prostate cancer progression. These adaptations are androgen-dependent and provide evidence for a novel hormone-regulated mechanism that support establishment and extension of MAMs. Future studies will determine whether these changes are required to maintain pro-proliferative signaling and metabolic changes that support prostate cancer cell viability.
Collapse
Affiliation(s)
- Lisa M. Butler
- South Australian Immunogenomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Emma Evergren
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
4
|
de Ridder I, Kerkhofs M, Lemos FO, Loncke J, Bultynck G, Parys JB. The ER-mitochondria interface, where Ca 2+ and cell death meet. Cell Calcium 2023; 112:102743. [PMID: 37126911 DOI: 10.1016/j.ceca.2023.102743] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/03/2023]
Abstract
Endoplasmic reticulum (ER)-mitochondria contact sites are crucial to allow Ca2+ flux between them and a plethora of proteins participate in tethering both organelles together. Inositol 1,4,5-trisphosphate receptors (IP3Rs) play a pivotal role at such contact sites, participating in both ER-mitochondria tethering and as Ca2+-transport system that delivers Ca2+ from the ER towards mitochondria. At the ER-mitochondria contact sites, the IP3Rs function as a multi-protein complex linked to the voltage-dependent anion channel 1 (VDAC1) in the outer mitochondrial membrane, via the chaperone glucose-regulated protein 75 (GRP75). This IP3R-GRP75-VDAC1 complex supports the efficient transfer of Ca2+ from the ER into the mitochondrial intermembrane space, from which the Ca2+ ions can reach the mitochondrial matrix through the mitochondrial calcium uniporter. Under physiological conditions, basal Ca2+ oscillations deliver Ca2+ to the mitochondrial matrix, thereby stimulating mitochondrial oxidative metabolism. However, when mitochondrial Ca2+ overload occurs, the increase in [Ca2+] will induce the opening of the mitochondrial permeability transition pore, thereby provoking cell death. The IP3R-GRP75-VDAC1 complex forms a hub for several other proteins that stabilize the complex and/or regulate the complex's ability to channel Ca2+ into the mitochondria. These proteins and their mechanisms of action are discussed in the present review with special attention for their role in pathological conditions and potential implication for therapeutic strategies.
Collapse
Affiliation(s)
- Ian de Ridder
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium
| | - Martijn Kerkhofs
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium
| | - Fernanda O Lemos
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium
| | - Jens Loncke
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium.
| | - Jan B Parys
- KU Leuven, Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, Leuven BE-3000, Belgium.
| |
Collapse
|
5
|
Key genes expressed in mitochondria‑endoplasmic reticulum contact sites in cancer (Review). Oncol Rep 2023; 49:77. [PMID: 36866764 PMCID: PMC10018458 DOI: 10.3892/or.2023.8514] [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: 08/05/2022] [Accepted: 12/06/2022] [Indexed: 03/04/2023] Open
Abstract
Cell fate is critically affected by mitochondrial activity, from ATP production to metabolism, Ca2+ homeostasis and signaling. These actions are regulated by proteins expressed in mitochondria (Mt)‑endoplasmic reticulum contact sites (MERCSs). The literature supports the fact that disruption to the physiology of the Mt and/or MERCSs can be due to alterations in the Ca2+ influx/efflux, which further regulates autophagy and apoptosis activity. The current review presents the findings of numerous studies with regard to the involvement of proteins positioned in MERCSs and how they express anti‑ and pro‑apoptotic properties by adjusting Ca2+ across membranes. The review also explores the involvement of mitochondrial proteins as hot spots in cancer development, cell death and/or survival, and the method via which they can potentially be targeted as a therapeutic option.
Collapse
|
6
|
Marschner RA, Roginski AC, Ribeiro RT, Longo L, Álvares-da-Silva MR, Wajner SM. Uncovering Actions of Type 3 Deiodinase in the Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). Cells 2023; 12:cells12071022. [PMID: 37048095 PMCID: PMC10093729 DOI: 10.3390/cells12071022] [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: 12/26/2022] [Revised: 02/16/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has gained worldwide attention as a public health problem. Nonetheless, lack of enough mechanistic knowledge restrains effective treatments. It is known that thyroid hormone triiodothyronine (T3) regulates hepatic lipid metabolism, and mitochondrial function. Liver dysfunction of type 3 deiodinase (D3) contributes to MAFLD, but its role is not fully understood. Objective: To evaluate the role of D3 in the progression of MAFLD in an animal model. Methodology: Male/adult Sprague Dawley rats (n = 20) were allocated to a control group (2.93 kcal/g) and high-fat diet group (4.3 kcal/g). Euthanasia took place on the 28th week. D3 activity and expression, Uncoupling Protein 2 (UCP2) and type 1 deiodinase (D1) expression, oxidative stress status, mitochondrial, Krebs cycle and endoplasmic reticulum homeostasis in liver tissue were measured. Results: We observed an increase in D3 activity/expression (p < 0.001) related to increased thiobarbituric acid reactive substances (TBARS) and carbonyls and diminished reduced glutathione (GSH) in the MAFLD group (p < 0.05). There was a D3-dependent decrease in UCP2 expression (p = 0.01), mitochondrial capacity, respiratory activity with increased endoplasmic reticulum stress in the MAFLD group (p < 0.001). Surprisingly, in an environment with lower T3 levels due to high D3 activity, we observed an augmented alpha-ketoglutarate dehydrogenase (KGDH) and glutamate dehydrogenase (GDH) enzymes activity (p < 0.05). Conclusion: Induced D3, triggered by changes in the REDOX state, decreases T3 availability and hepatic mitochondrial capacity. The Krebs cycle enzymes were altered as well as endoplasmic reticulum stress. Taken together, these results shed new light on the role of D3 metabolism in MAFLD.
Collapse
Affiliation(s)
- Rafael Aguiar Marschner
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
| | - Ana Cristina Roginski
- Post-Graduate Program in Biochemestry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, RS, Brazil
| | - Rafael Teixeira Ribeiro
- Post-Graduate Program in Biochemestry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, RS, Brazil
| | - Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Center for Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, RS, Brazil
- Department of Internal Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, RS, Brazil
| | - Simone Magagnin Wajner
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil
- Department of Internal Medicine, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, RS, Brazil
- Correspondence:
| |
Collapse
|
7
|
Ganji C, Muppala V, Khan M, Purnachandra Nagaraju G, Farran B. Mitochondrial-targeted nanoparticles: Delivery and therapeutic agents in cancer. Drug Discov Today 2023; 28:103469. [PMID: 36529353 DOI: 10.1016/j.drudis.2022.103469] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Mitochondria are the powerhouses of cells and modulate the essential metabolic functions required for cellular survival. Various mitochondrial pathways, such as oxidative phosphorylation or production of reactive oxygen species (ROS) are dysregulated during cancer growth and development, rendering them attractive targets against cancer. Thus, the delivery of antitumor agents to mitochondria has emerged as a potential approach for treating cancer. Recent advances in nanotechnology have provided innovative solutions for overcoming the physical barriers posed by the structure of mitochondrial organelles, and have enabled the development of efficient mitochondrial nanoplatforms. In this review, we examine the importance of mitochondria during neoplastic development, explore the most recent smart designs of nano-based systems aimed at targeting mitochondria, and highlight key mitochondrial pathways in cancer cells.
Collapse
Affiliation(s)
- Chaithanya Ganji
- Department of Hematology and Oncology, School of Medicine, University of Alabama, Birmingham, AL 35201, USA
| | - Veda Muppala
- Department of Hematology and Oncology, School of Medicine, University of Alabama, Birmingham, AL 35201, USA
| | - Musaab Khan
- Department of Hematology and Oncology, School of Medicine, University of Alabama, Birmingham, AL 35201, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama, Birmingham, AL 35201, USA.
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
8
|
Wei W, Lu H, Dai W, Zheng X, Dong H. Multiplexed Organelles Portrait Barcodes for Subcellular MicroRNA Array Detection in Living Cells. ACS NANO 2022; 16:20329-20339. [PMID: 36410732 DOI: 10.1021/acsnano.2c06252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Multiplexed profiling of microRNAs' subcellular expression and distribution is essential to understand their spatiotemporal function information, but it remains a crucial challenge. Herein, we report an encoding approach that leverages combinational fluorescent dye barcodes, organelle targeting elements, and an independent quantification signal, termed Multiplexed Organelles Portrait Barcodes (MOPB), for high-throughput profiling of miRNAs from organelles. The MOPB barcodes consist of heterochromatic fluorescent dye-loaded shell-core mesoporous silica nanoparticles modified with organelle targeting peptides and molecular beacon detection probes. Using mitochondria and endoplasmic reticulum as models, we encoded four Cy3/AMCA ER-MOPB and four Cy5/AMCA Mito-MOPB by varying the Cy3 and Cy5 intensity for distinguishing eight organelles' miRNAs. Significantly, the MOPB strategy successfully and accurately profiled eight subcellular organelle miRNAs' alterations in the drug-induced Ca2+ homeostasis breakdown. The approach should allow more widespread application of subcellular miRNAs and multiplexed subcellular protein biomarkers' monitoring for drug discovery, cellular metabolism, signaling transduction, and gene expression regulation readout.
Collapse
Affiliation(s)
- Wei Wei
- Marshall Laboratory of Biomedical Engineering, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University, 3688 Nanhai Road, Shenzhen, Guangdong518060, China
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Bioengineering, University of Science and Technology Beijing30 Xueyuan Road, 100083, Beijing, China
| | - Huiting Lu
- Department of Chemistry, School of Chemistry and Bioengineering, University of Science and Technology Beijing, 30 Xueyuan Road, Beijing100083, China
| | - Wenhao Dai
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Bioengineering, University of Science and Technology Beijing30 Xueyuan Road, 100083, Beijing, China
| | - Xiaonan Zheng
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Bioengineering, University of Science and Technology Beijing30 Xueyuan Road, 100083, Beijing, China
| | - Haifeng Dong
- Marshall Laboratory of Biomedical Engineering, Research Center for Biosensor and Nanotheranostic, School of Biomedical Engineering, Shenzhen University, 3688 Nanhai Road, Shenzhen, Guangdong518060, China
| |
Collapse
|
9
|
Luby A, Alves-Guerra MC. UCP2 as a Cancer Target through Energy Metabolism and Oxidative Stress Control. Int J Mol Sci 2022; 23:ijms232315077. [PMID: 36499405 PMCID: PMC9735768 DOI: 10.3390/ijms232315077] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/02/2022] Open
Abstract
Despite numerous therapies, cancer remains one of the leading causes of death worldwide due to the lack of markers for early detection and response to treatment in many patients. Technological advances in tumor screening and renewed interest in energy metabolism have allowed us to identify new cellular players in order to develop personalized treatments. Among the metabolic actors, the mitochondrial transporter uncoupling protein 2 (UCP2), whose expression is increased in many cancers, has been identified as an interesting target in tumor metabolic reprogramming. Over the past decade, a better understanding of its biochemical and physiological functions has established a role for UCP2 in (1) protecting cells from oxidative stress, (2) regulating tumor progression through changes in glycolytic, oxidative and calcium metabolism, and (3) increasing antitumor immunity in the tumor microenvironment to limit cancer development. With these pleiotropic roles, UCP2 can be considered as a potential tumor biomarker that may be interesting to target positively or negatively, depending on the type, metabolic status and stage of tumors, in combination with conventional chemotherapy or immunotherapy to control tumor development and increase response to treatment. This review provides an overview of the latest published science linking mitochondrial UCP2 activity to the tumor context.
Collapse
|
10
|
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
|
11
|
Albaradei S, Albaradei A, Alsaedi A, Uludag M, Thafar MA, Gojobori T, Essack M, Gao X. MetastaSite: Predicting metastasis to different sites using deep learning with gene expression data. Front Mol Biosci 2022; 9:913602. [PMID: 35936793 PMCID: PMC9353773 DOI: 10.3389/fmolb.2022.913602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/29/2022] [Indexed: 12/03/2022] Open
Abstract
Deep learning has massive potential in predicting phenotype from different omics profiles. However, deep neural networks are viewed as black boxes, providing predictions without explanation. Therefore, the requirements for these models to become interpretable are increasing, especially in the medical field. Here we propose a computational framework that takes the gene expression profile of any primary cancer sample and predicts whether patients' samples are primary (localized) or metastasized to the brain, bone, lung, or liver based on deep learning architecture. Specifically, we first constructed an AutoEncoder framework to learn the non-linear relationship between genes, and then DeepLIFT was applied to calculate genes' importance scores. Next, to mine the top essential genes that can distinguish the primary and metastasized tumors, we iteratively added ten top-ranked genes based upon their importance score to train a DNN model. Then we trained a final multi-class DNN that uses the output from the previous part as an input and predicts whether samples are primary or metastasized to the brain, bone, lung, or liver. The prediction performances ranged from AUC of 0.93-0.82. We further designed the model's workflow to provide a second functionality beyond metastasis site prediction, i.e., to identify the biological functions that the DL model uses to perform the prediction. To our knowledge, this is the first multi-class DNN model developed for the generic prediction of metastasis to various sites.
Collapse
Affiliation(s)
- Somayah Albaradei
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Asim Alsaedi
- King Saud Bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Mahmut Uludag
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Maha A. Thafar
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- College of Computers and Information Technology, Taif University, Taif, Saudi Arabia
| | - Takashi Gojobori
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Magbubah Essack
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computer Electrical and Mathematical Sciences and Engineering Division (CEMSE), Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
12
|
Li J, Qi F, Su H, Zhang C, Zhang Q, Chen Y, Chen P, Su L, Chen Y, Yang Y, Chen Z, Zhang S. GRP75-faciliated Mitochondria-associated ER Membrane (MAM) Integrity controls Cisplatin-resistance in Ovarian Cancer Patients. Int J Biol Sci 2022; 18:2914-2931. [PMID: 35541901 PMCID: PMC9066115 DOI: 10.7150/ijbs.71571] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/01/2022] [Indexed: 11/19/2022] Open
Abstract
Background: Control of ER-mitochondrial Ca2+ fluxes is a critical checkpoint to determine cell fate under stress. The 75-kDa glucose-regulated protein (GRP75) is a key tether protein facilitating mitochondria-associated ER membrane (MAM) formation through the IP3R-GRP75-VDAC1 complex. Although GRP75 contributes to cisplatin (CP)-resistance of ovarian cancer (OC), the underlying mechanisms are not clear. Methods: CP-resistant and -sensitive OC cell lines with GRP75 stable modulation were established. Confocal, PLA, co-IP, and TEM analysis were utilized to detect MAM integrity. Live cell Ca2+ imaging, intracellular ATP, ROS, and NAD+ assays were utilized to investigate ER-to-mitochondrial Ca2+ transfer and mitochondrial bioenergetics. Western blot, flow cytometry, CCK-8, Δψm, and mPTP assays were utilized to examine apoptotic cell death. Bioinformatics, patient's specimens, and immunohistochemistry were conducted to obtain the clinical relevance for GRP75-facilitated MAM formation. Results: GRP75-faciliated MAM formation was enriched in CP-resistant OC cells. CP-exposure only increased MAM formation in CP-sensitive OC cells, and enrichment of GRP75 and VDAC1 at MAMs is indispensable to CP-resistance. Diminishing MAM integrity by GRP75-deficiency reduced ER-to-mitochondria Ca2+ transfer, accelerated CP-induced mitochondrial dysfunction, provoked catastrophic ROS, and enhanced CP-triggered apoptotic cell death in OC cells. Clinical investigations confirmed the enrichment of GRP75-faciliated MAM formation in relapsed OC patients, and such enrichment was associated with the CP-resistance phenotype. Conclusion: GRP75-overexpression confers CP-resistance by distinctively managing MAM-facilitated Ca2+ fluxes and the pro-survival ROS signal, whereas GRP75-deficiency induces cell death via bioenergetic crisis and apoptotic ROS accumulation in OC cells. Our results show that GRP75-faciliated MAM formation is a potential target to overcome CP-resistance of OC.
Collapse
Affiliation(s)
- Jing Li
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Fangzheng Qi
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Huishan Su
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Chuanshan Zhang
- Department of Pathology, Third Central Hospital of Tianjin Medical University, 83 Jintang Road, Tianjin, 300170, P. R. China
| | - Qing Zhang
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Ying Chen
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Ping Chen
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Linjia Su
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Yanan Chen
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Zhesheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York, NY, 11439, USA
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
13
|
Tawfik I, Gottschalk B, Jarc A, Bresilla D, Rost R, Obermayer-Pietsch B, Graier WF, Madreiter-Sokolowski CT. T3-induced enhancement of mitochondrial Ca 2+ uptake as a boost for mitochondrial metabolism. Free Radic Biol Med 2022; 181:197-208. [PMID: 35091061 DOI: 10.1016/j.freeradbiomed.2022.01.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022]
Abstract
Thyroid hormones act as master regulators of cellular metabolism. Thereby, the biologically active triiodothyronine (T3) induces the expression of genes to enhance mitochondrial metabolic function. Notably, Ca2+ ions are necessary for the activity of dehydrogenases of the tricarboxylic acid cycle and, thus, mitochondrial respiration. We investigated whether treating HeLa cells with T3 causes alterations in mitochondrial Ca2+ ([Ca2+]mito) levels. Real-time measurements by fluorescence microscopy revealed that treatment with T3 for 3 h induces a significant increase in basal [Ca2+]mito levels and [Ca2+]mito uptake upon the depletion of the endoplasmic reticulum (ER) Ca2+ store, while cytosolic Ca2+ levels remained unchanged. T3 incubation was found to upregulate mRNA expression levels of uncoupling proteins 2 and 3 (UCP2, UCP3) and of protein arginine methyltransferase 1 (PRMT1). Live-cell imaging revealed that T3-induced enhancement of mitochondrial Ca2+ uptake depends on the mitochondrial Ca2+ uniporter (MCU), UCP2, and PRMT1 that are essential for increased mitochondrial ATP ([ATP]mito) production after T3 treatment. Besides, increased [Ca2+]mito and [ATP]mito levels correlated with enhanced production of reactive oxygen species (ROS) in mitochondria. Notably, ROS scavenging causes mitochondrial Ca2+ elevation and outplays the impact of T3 on [Ca2+]mito homeostasis. Based on these results, we assume that thyroid hormones adjust [Ca2+]mito homeostasis by modulating the UCP2- and PRMT1-balanced [Ca2+]mito uptake via MCU in case of physiological ROS levels to convey their impact on mitochondrial ATP and ROS production.
Collapse
Affiliation(s)
- Ines Tawfik
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Angelo Jarc
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Doruntina Bresilla
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Barbara Obermayer-Pietsch
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrinology Lab Platform, Medical University of Graz, Auenbruggerplatz 15, 8010, Graz, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria; BioTechMed, Graz, Austria
| | - Corina T Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
| |
Collapse
|
14
|
Koshenov Z, Oflaz FE, Hirtl M, Gottschalk B, Rost R, Malli R, Graier WF. Citrin mediated metabolic rewiring in response to altered basal subcellular Ca 2+ homeostasis. Commun Biol 2022; 5:76. [PMID: 35058562 PMCID: PMC8776887 DOI: 10.1038/s42003-022-03019-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/28/2021] [Indexed: 01/19/2023] Open
Abstract
In contrast to long-term metabolic reprogramming, metabolic rewiring represents an instant and reversible cellular adaptation to physiological or pathological stress. Ca2+ signals of distinct spatio-temporal patterns control a plethora of signaling processes and can determine basal cellular metabolic setting, however, Ca2+ signals that define metabolic rewiring have not been conclusively identified and characterized. Here, we reveal the existence of a basal Ca2+ flux originating from extracellular space and delivered to mitochondria by Ca2+ leakage from inositol triphosphate receptors in mitochondria-associated membranes. This Ca2+ flux primes mitochondrial metabolism by maintaining glycolysis and keeping mitochondria energized for ATP production. We identified citrin, a well-defined Ca2+-binding component of malate-aspartate shuttle in the mitochondrial intermembrane space, as predominant target of this basal Ca2+ regulation. Our data emphasize that any manipulation of this ubiquitous Ca2+ system has the potency to initiate metabolic rewiring as an instant and reversible cellular adaptation to physiological or pathological stress.
Collapse
Affiliation(s)
- Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Furkan E Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria
- BioTechMed Graz, 8010, Graz, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010, Graz, Austria.
- BioTechMed Graz, 8010, Graz, Austria.
| |
Collapse
|
15
|
Gottschalk B, Madreiter-Sokolowski CT, Graier WF. Cristae junction as a fundamental switchboard for mitochondrial ion signaling and bioenergetics. Cell Calcium 2022; 101:102517. [PMID: 34915234 DOI: 10.1016/j.ceca.2021.102517] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/08/2021] [Indexed: 12/31/2022]
Abstract
OPA1 and MICU1 are both involved in the regulation of mitochondrial Ca2+ uptake and the stabilization of the cristae junction, which separates the inner mitochondrial membrane into the interboundary membrane and the cristae membrane. In this mini-review, we focus on the synergetic control of OPA1 and MICU1 on the cristae junction that serves as a fundamental regulator of multiple mitochondrial functions. In particular, we point to the critical role of an adaptive cristae junction permeability in mitochondrial Ca2+ signaling, spatial H+ gradients and mitochondrial membrane potential, metabolic activity, and apoptosis. These characteristics bear on a distinct localization of the oxidative phosphorylation machinery, the FoF1-ATPase, and mitochondrial Ca2+uniporter (MCU) within sections of the inner mitochondrial membrane isolated by the cristae junction and regulated by proteins like OPA1 and MICU1. We specifically focus on the impact of MICU1-regulated cristae junction on the activity and distribution of MCU within the complex ultrastructure of mitochondria.
Collapse
Affiliation(s)
- Benjamin Gottschalk
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, Graz, 8010 Austria
| | - Corina T Madreiter-Sokolowski
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, Graz, 8010 Austria
| | - Wolfgang F Graier
- Gottfried Schatz Research Center: Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/6, Graz, 8010 Austria; BioTechMed, Graz.
| |
Collapse
|
16
|
Hu T, Qin Z, Shen C, Gong HL, He ZY. Multifunctional Mitochondria-Targeting Nanosystems for Enhanced Anticancer Efficacy. Front Bioeng Biotechnol 2021; 9:786621. [PMID: 34900973 PMCID: PMC8652136 DOI: 10.3389/fbioe.2021.786621] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 02/05/2023] Open
Abstract
Mitochondria, a kind of subcellular organelle, play crucial roles in cancer cells as an energy source and as a generator of reactive substrates, which concern the generation, proliferation, drug resistance, and other functions of cancer. Therefore, precise delivery of anticancer agents to mitochondria can be a novel strategy for enhanced cancer treatment. Mitochondria have a four-layer structure with a high negative potential, which thereby prevents many molecules from reaching the mitochondria. Luckily, the advances in nanosystems have provided enormous hope to overcome this challenge. These nanosystems include liposomes, nanoparticles, and nanomicelles. Here, we summarize the very latest developments in mitochondria-targeting nanomedicines in cancer treatment as well as focus on designing multifunctional mitochondria-targeting nanosystems based on the latest nanotechnology.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Shen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Han-Lin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Yao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Devi RV, Raj D, Doble M. Lockdown of mitochondrial Ca 2+ extrusion and subsequent resveratrol treatment kill HeLa cells by Ca 2+ overload. Int J Biochem Cell Biol 2021; 139:106071. [PMID: 34428589 DOI: 10.1016/j.biocel.2021.106071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/06/2021] [Accepted: 08/19/2021] [Indexed: 10/20/2022]
Abstract
Anticancer effect of resveratrol and the role of sodium/lithium/calcium exchanger in context with calcium ions are studied in human cervical cancer cell line. This therapeutic approach using siNCLX mediated gene silencing and drug therapy with resveratrol indicates the disruption of calcium homeostasis, increase in caspase (-3, 8, 9) mRNA expressions and DNA damage leading to apoptotic cell death. Monitoring the intracellular Ca2+ changes using fluo-4AM indicates highest rise in [Ca2+] level in sodium/lithium/calcium exchanger silenced group with five different stages, that is distinguishable based on the fluorescence intensity. In resveratrol treated and siNCLX + resveratrol treated groups no such cell staging differences were observed, despite uniform Ca2+ rise followed by decrease in the intensity. Integrating RNAi gene silencing of sodium/lithium/calcium exchanger with resveratrol can form the most interesting, efficient and promising therapeutic strategy in the treatment of cancer.
Collapse
Affiliation(s)
- R Viswambari Devi
- Bioengineering and Drug Design Laboratory, Department of Biotechnology, Indian Institute of Technology, Chennai, 600036, India
| | - Divakar Raj
- Bioengineering and Drug Design Laboratory, Department of Biotechnology, Indian Institute of Technology, Chennai, 600036, India
| | - Mukesh Doble
- Bioengineering and Drug Design Laboratory, Department of Biotechnology, Indian Institute of Technology, Chennai, 600036, India.
| |
Collapse
|
18
|
Oflaz FE, Koshenov Z, Hirtl M, Rost R, Bachkoenig OA, Gottschalk B, Madreiter-Sokolowski CT, Malli R, Graier WF. Near-UV Light Induced ROS Production Initiates Spatial Ca 2+ Spiking to Fire NFATc3 Translocation. Int J Mol Sci 2021; 22:8189. [PMID: 34360954 PMCID: PMC8346968 DOI: 10.3390/ijms22158189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 01/19/2023] Open
Abstract
Ca2+-dependent gene regulation controls several functions to determine the fate of the cells. Proteins of the nuclear factor of activated T-cells (NFAT) family are Ca2+ sensitive transcription factors that control the cell growth, proliferation and insulin secretion in β-cells. Translocation of NFAT proteins to the nucleus occurs in a sequence of events that starts with activating calmodulin-dependent phosphatase calcineurin in a Ca2+-dependent manner, which dephosphorylates the NFAT proteins and leads to their translocation to the nucleus. Here, we examined the role of IP3-generating agonists and near-UV light in the induction of NFATc3 migration to the nucleus in the pancreatic β-cell line INS-1. Our results show that IP3 generation yields cytosolic Ca2+ rise and NFATc3 translocation. Moreover, near-UV light exposure generates reactive oxygen species (ROS), resulting in cytosolic Ca2+ spiking via the L-type Ca2+ channel and triggers NFATc3 translocation to the nucleus. Using the mitochondria as a Ca2+ buffering tool, we showed that ROS-induced cytosolic Ca2+ spiking, not the ROS themselves, was the triggering mechanism of nuclear import of NFATc3. Collectively, this study reveals the mechanism of near-UV light induced NFATc3 migration.
Collapse
Affiliation(s)
- Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Corina T. Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
19
|
Koshenov Z, Oflaz FE, Hirtl M, Pilic J, Bachkoenig OA, Gottschalk B, Madreiter-Sokolowski CT, Rost R, Malli R, Graier WF. Sigma-1 Receptor Promotes Mitochondrial Bioenergetics by Orchestrating ER Ca 2+ Leak during Early ER Stress. Metabolites 2021; 11:422. [PMID: 34206832 PMCID: PMC8305890 DOI: 10.3390/metabo11070422] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/11/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
The endoplasmic reticulum (ER) is a complex, multifunctional organelle of eukaryotic cells and responsible for the trafficking and processing of nearly 30% of all human proteins. Any disturbance to these processes can cause ER stress, which initiates an adaptive mechanism called unfolded protein response (UPR) to restore ER functions and homeostasis. Mitochondrial ATP production is necessary to meet the high energy demand of the UPR, while the molecular mechanisms of ER to mitochondria crosstalk under such stress conditions remain mainly enigmatic. Thus, better understanding the regulation of mitochondrial bioenergetics during ER stress is essential to combat many pathologies involving ER stress, the UPR, and mitochondria. This article investigates the role of Sigma-1 Receptor (S1R), an ER chaperone, has in enhancing mitochondrial bioenergetics during early ER stress using human neuroblastoma cell lines. Our results show that inducing ER stress with tunicamycin, a known ER stressor, greatly enhances mitochondrial bioenergetics in a time- and S1R-dependent manner. This is achieved by enhanced ER Ca2+ leak directed towards mitochondria by S1R during the early phase of ER stress. Our data point to the importance of S1R in promoting mitochondrial bioenergetics and maintaining balanced H2O2 metabolism during early ER stress.
Collapse
Affiliation(s)
- Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Johannes Pilic
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Corina T. Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (Z.K.); (F.E.O.); (M.H.); (J.P.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.R.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|