1
|
Dewan S, Sonker H, Chaudhary K, Agrawal S, Chaudhary A, Kumar A, Agrahari B, Singh RG. Self-Assembling Imidazolium Nanoaggregates Trigger a Unique Dynamin-Dependent Cell Death via Cytoplasmic Vacuolization and Mitochondrial Dysfunction in Human Lung Adenocarcinoma. J Med Chem 2025. [PMID: 40408548 DOI: 10.1021/acs.jmedchem.5c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
The identification of alternative cell death pathways is key to developing therapies for apoptosis-resistant cancers. We investigated cell death induced by delocalized lipophilic cation (DLC) nanoaggregates in A549 lung carcinoma cells. These DLCs trigger a dynamin-dependent, nonapoptotic pathway involving cytoplasmic vesicle accumulation and mitochondrial dysfunction. Leveraging the mitochondria-targeting ability of lipophilic cations, we designed and synthesized fluorescent mitochondrion-toxic molecules with potent cytotoxicity against A549, MDA-MB-231, and MCF-7 cells. Dynamic light scattering revealed the nanoaggregate formation of the lead compound, L3, in the RPMI media. L3 inhibited metastasis and clonal expansion, induced vacuole formation post endocytosis, and impaired the mitochondrial function, disrupting ATP levels. This led to mitochondrial permeability transition pore (MPTP) opening and oxidative imbalance via glutathione perturbation. L3 demonstrated strong antitumor activity in vitro and in vivo, showing high potential for treating apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Sayari Dewan
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Himanshu Sonker
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Kajal Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Saloni Agrawal
- Department of Biological Science and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ayushi Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashwini Kumar
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Bhumika Agrahari
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ritika Gautam Singh
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, India
| |
Collapse
|
2
|
Oliveira GL, Novak J, Nahacka Z, Brisudova P, Mota SI, Neuzil J, Oliveira PJ, Marques R. Adenine nucleotide translocase 2 silencing promotes metabolic reprogramming in P19 embryonal carcinoma stem cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167902. [PMID: 40381816 DOI: 10.1016/j.bbadis.2025.167902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 04/22/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Although controversial, cancer stem cells (CSCs) are thought to be one tumor component, being characterized by their strong self-renewal and survival properties. Cancer cells, CSCs included, are thought to rely mostly on glycolysis, even in the presence of oxygen, which confers them adaptive advantages. Adenine nucleotide translocator 2 (ANT2), responsible for the exchange of ADP and ATP in the mitochondrial inner membrane, has been correlated with a higher glycolytic metabolism and is known to be overexpressed in cancer cells. Using P19 embryonal carcinoma stem cells, we inhibited ANT2 translation by using siRNA. ANT2 protein levels were shown to be overexpressed in P19 undifferentiated cells (P19SCs) when compared to their differentiated counterparts (P19dCs). Furthermore, we showed here that the OXPHOS machinery and mitochondrial membrane potential are compromised after ANT2 depletion, leading to a metabolic adaptation towards a less oxidative phenotype. Interestingly, hexokinase II levels were downregulated, which was also accompanied by decreased cell growth, and reduced ability to form spheroids. Our findings underscore ANT2 as a key regulator of metabolic remodeling and cell survival of cancer stem-like cells, suggesting its potential as a therapeutic target for controlling CSC-driven tumor progression.
Collapse
Affiliation(s)
- Gabriela L Oliveira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; III - Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789 Coimbra, Portugal; Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jaromir Novak
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Petra Brisudova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Sandra I Mota
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; III - Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic; School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Paulo J Oliveira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| | - Ricardo Marques
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal; BRIDGES - Biotechnology Research, Innovation and Design for Health Products, Polytechnic University of Guarda, Guarda, Portugal.
| |
Collapse
|
3
|
Yosef O, Cohen-Daniel L, Shamriz O, Bar-On Z, Salaymeh W, Saragovi A, Abramovich I, Agranovich B, Lutz V, Tam J, Permyakova A, Gottlieb E, Huber M, Berger M. Metabolic reprogramming driven by Ant2 deficiency augments T Cell function and anti-tumor immunity in mice. Nat Commun 2025; 16:4292. [PMID: 40341170 PMCID: PMC12062294 DOI: 10.1038/s41467-025-59310-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
T cell activation requires a substantial increase in NAD+ production, often exceeding the capacity of oxidative phosphorylation (OXPHOS). To investigate how T cells adapt to this metabolic challenge, we generate T cell-specific ADP/ATP translocase-2 knockout (Ant2-/-) mice. Loss of Ant2, a crucial protein mediating ADP/ATP exchange between mitochondria and cytoplasm, induces OXPHOS restriction by limiting ATP synthase activity, thereby impeding NAD+ regeneration. Interestingly, Ant2-/- naïve T cells exhibit enhanced activation, proliferation and effector functions compared to wild-type controls. Metabolic profiling reveals that these T cells adopt an activated-like metabolic program with increased mitobiogenesis and anabolism. Lastly, pharmacological inhibition of ANT in wild-type T cells recapitulates the Ant2-/- phenotype and improves adoptive T cell therapy of cancer in mouse models. Our findings thus suggest that Ant2-deficient T cells bypass the typical metabolic reprogramming required for activation, leading to enhanced T cell function and highlighting the therapeutic potential of targeting ANT for immune modulation.
Collapse
Affiliation(s)
- Omri Yosef
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leonor Cohen-Daniel
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oded Shamriz
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zahala Bar-On
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeeh Salaymeh
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amijai Saragovi
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Permyakova
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Gottlieb
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Michael Berger
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
4
|
Bohacova K, Nahacka Z, Dudova J, Kovarova J, Rohlena J, Rennerova M, Kasperova BJ, Stursa J, Werner L, Haluzik M, Neuzil J, Hubackova SS. Role of ANT2 in mitochondrial function and cancer cell survival: a target for therapeutic intervention. Cell Death Discov 2025; 11:225. [PMID: 40335504 PMCID: PMC12059193 DOI: 10.1038/s41420-025-02510-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/04/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025] Open
Affiliation(s)
- Klara Bohacova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia.
- Faculty of Science, Charles University, Prague, Czechia.
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia.
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
| | - Jana Dudova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Czechia
| | - Jaromira Kovarova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
| | - Michaela Rennerova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Barbora Judita Kasperova
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
- First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Jan Stursa
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Lukas Werner
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia
- Faculty of Science, Charles University, Prague, Czechia
- First Faculty of Medicine, Charles University in Prague, Prague, Czechia
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, Australia
| | - Sona Stemberkova Hubackova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czechia.
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia.
| |
Collapse
|
5
|
Yamada A, Watanabe A, Nara A, Inokuma T, Asano M, Shinohara Y, Yamamoto T. Multiple Inhibitory Mechanisms of DS16570511 Targeting Mitochondrial Calcium Uptake: Insights from Biochemical Analysis of Rat Liver Mitochondria. Int J Mol Sci 2025; 26:2670. [PMID: 40141312 PMCID: PMC11942279 DOI: 10.3390/ijms26062670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Mitochondrial calcium (Ca2+) uptake plays a key role in mitochondrial physiology and disease development. This process is regulated by the mitochondrial calcium uniporter (MCU) complex. DS16570511 is a membrane-permeable drug that inhibits mitochondrial Ca2+ uptake, although its inhibitory mechanisms remain unclear. In this study, we evaluated the effects of DS16570511 on various mitochondrial functions through biochemical analyses. We found that DS16570511 affects multiple mitochondrial functions and exhibits variable potency in inhibiting individual processes. Specifically, DS16570511 not only inhibits MCU, its initially reported target, but also respiratory chain complexes and FoF1-adenosine triphosphatase/adenine nucleotide translocator, particularly respiratory chain complex II. Furthermore, the carboxyl group at the molecular terminus of DS16570511 plays a critical role in its inhibitory effects on mitochondrial Ca2+ uptake through respiratory chain complex II inhibition. These findings enhance our understanding of the mechanisms by which DS16570511 inhibits mitochondrial Ca2+ uptake and provide valuable insights for the clinical application of mitochondrial Ca2+ uptake inhibitors.
Collapse
Affiliation(s)
- Akiko Yamada
- Department of Pathology, Nihon University School of Dentistry, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Akira Watanabe
- Institute for Genome Research, Tokushima University, Tokushima 770-8503, Tokushima, Japan
- Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8503, Tokushima, Japan
| | - Atsushi Nara
- Institute for Genome Research, Tokushima University, Tokushima 770-8503, Tokushima, Japan
- Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8503, Tokushima, Japan
| | - Tsubasa Inokuma
- Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8503, Tokushima, Japan
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, Chiyoda-ku, Tokyo 101-8310, Japan
| | - Yasuo Shinohara
- Institute for Genome Research, Tokushima University, Tokushima 770-8503, Tokushima, Japan
- Graduate School of Pharmaceutical Sciences, Tokushima University, Tokushima 770-8503, Tokushima, Japan
| | - Takenori Yamamoto
- Institute for Genome Research, Tokushima University, Tokushima 770-8503, Tokushima, Japan
- Division of Molecular Target and Gene Therapy Products, National Institute of Health Sciences, Kawasaki 210-9501, Kanagawa, Japan
| |
Collapse
|
6
|
Pohl EE, Vazdar M, Kreiter J. Exploring the proton transport mechanism of the mitochondrial ADP/ATP carrier: FA-cycling hypothesis and beyond. Protein Sci 2025; 34:e70047. [PMID: 39969060 PMCID: PMC11837047 DOI: 10.1002/pro.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/20/2025]
Abstract
The mitochondrial ADP/ATP carrier (AAC, ANT), a member of the SLC25 family of solute carriers, plays a critical role in transporting purine nucleotides (ATP and ADP) as well as protons across the inner mitochondrial membrane. However, the precise mechanism and physiological significance of proton transport by ADP/ATP carrier remain unclear. Notably, the presence of uncouplers-such as long-chain fatty acids (FA) or artificial compounds like dinitrophenol (DNP)-is essential for this process. We explore two potential mechanisms that describe ADP/ATP carrier as either (i) a proton carrier that functions in the presence of FA or DNP, or (ii) an anion transporter (FA- or DNP). In the latter case, the proton is translocated by the neutral form of FA, which carries it from the matrix to the intermembrane space (FA-cycling hypothesis). Our recent results support this hypothesis. We describe a four-step mechanism for the "sliding" of the FA anion from the matrix to the mitochondrial intermembrane space and discuss a possible generalization of this mechanism to other SLC25 carriers.
Collapse
Affiliation(s)
- Elena E. Pohl
- Physiology and Biophysics Unit, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| | - Mario Vazdar
- Department of Mathematics, Informatics, and CyberneticsUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Jürgen Kreiter
- Physiology and Biophysics Unit, Department of Biological Sciences and PathobiologyUniversity of Veterinary MedicineViennaAustria
| |
Collapse
|
7
|
Wang X, Liu J, Durga L, Beeraka NM, Zhou R, Lu P, Song R, Sinelnikov MY, Chen K, Fan R, Zhao D. Recent Updates on the Efficacy of Mitocans in Photo/Radio-therapy for Targeting Metabolism in Chemo/Radio-resistant Cancers: Nanotherapeutics. Curr Med Chem 2025; 32:2156-2182. [PMID: 38018190 DOI: 10.2174/0109298673259347231019121757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/27/2023] [Accepted: 09/15/2023] [Indexed: 11/30/2023]
Abstract
Conventional therapeutic modalities against the cancers such as surgery, chemotherapy (CT) and radiotherapy (RT) have limited efficacy due to drug resistance, and adverse effects. Recent developments in nanoscience emphasized novel approaches to overcome the aforementioned limitations and subsequently improve overall clinical outcomes in cancer patients. Photodynamic therapy (PDT), photothermal therapy (PTT), and radiodynamic therapy (RDT) can be used as cancer treatments due to their high selectivity, low drug resistance, and low toxicity. Mitocans are the therapeutic molecules that can produce anti-cancer effects by modulating mitochondria functions and they have significant implications in cancer therapy. Mitochondria- targeted therapy is a promising strategy in cancer treatment as these organelles play a crucial function in the regulation of apoptosis and metabolism in tumor cells and are more vulnerable to hyperthermia and oxidative damage. The aim of this review is used to explore the targeting efficacy of mitocans in the nanotherapeutic formulation when combined with therapies like PDT, PTT, RDT. We searched several databases include Pubmed, relemed, scopus, google scholar, Embase and collected the related information to the efficacy of mitocans in nanotherapeutics when combined with photo-radiotherapy to target chemo/radio-resisant tumor cells. In this review, we vividly described research reports pertinent to the selective delivery of chemotherapy molecules into specific sub-organelles which can significantly improve the efficiency of cancer treatment by targeting tumor cell metabolism. Furthermore, the rational design, functionalization and application of various mitochondrial targeting units, including organic phosphine/sulfur salts, quaternary ammonium salts, transition metal complexes, and mitochondria-targeted cancer therapy such as PDT, PTT, RDT, and others were summarized. Mainly, the efficacy of these modalities against mtDNA and additional nanotherapeutic strategies with photosensitizers, or radiotherapy to target mitochondrial metabolism in tumor cells with chemo/radio-resistance were delineated. This review can benefit nanotechnologists, oncologists, and radiation oncologists to develop rational designs and application of novel mitochondrial targeting drugs mainly to target metabolism in chemo/radio-resistant cancer cells in cancer therapy.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lakshmi Durga
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Narasimha M Beeraka
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian, Federation (Sechenov University), Moscow, Russia
| | - Runze Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengwei Lu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruixia Song
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mikhail Y Sinelnikov
- Sinelab Biomedical Research Centre, Minnesota, 55905, USA
- Department of General Medicine, University of Rome, Tor Vergata, Via Cracovia, 50, 00133, Rome, Italy
- Research Institute of Human Morphology, Russian Scientific Center of Surgery, Moscow, 119991, Russia
| | - Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Di Zhao
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Giannotti L, Di Chiara Stanca B, Spedicato F, Vergara D, Stanca E, Damiano F, Siculella L. Exploring the Therapeutic Potential of Cannabidiol in U87MG Cells: Effects on Autophagy and NRF2 Pathway. Antioxidants (Basel) 2024; 14:18. [PMID: 39857352 PMCID: PMC11761945 DOI: 10.3390/antiox14010018] [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: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Cannabinoids include both endogenous endocannabinoids and exogenous phytocannabinoids, such as cannabidiol (CBD), and have potential as therapeutic agents in cancer treatment due to their selective anticancer activities. CBD exhibits both antioxidant and pro-oxidant effects depending on its concentration and cell types. These properties allow CBD to influence oxidative stress responses and potentially enhance the efficacy of antitumor therapies. In this study, we treated U87MG glioma cells with low dose (1 μM) CBD and evaluated its molecular effects. Our findings indicate that CBD reduced cell viability by 20% (p < 0.05) through the alteration of mitochondrial membrane potential. The alteration of redox status by CBD caused an attempt to rescue mitochondrial functionality through nuclear localization of the GABP transcription factor involved in mitochondria biogenesis. Moreover, CBD treatment caused an increase in autophagic flux, as supported by the increase in Beclin-1 and the ratio of LC3-II/LC3-I. Due to mitochondria functionality alteration, pro-apoptotic proteins were induced without activating apoptotic effectors Caspase-3 or Caspase-7. The study of the transcription factor NRF2 and the ubiquitin-binding protein p62 expression revealed an increase in their levels in CBD-treated cells. In conclusion, low-dose CBD makes U87MG cells more vulnerable to cytotoxic effects, reducing cell viability and mitochondrial dynamics while increasing autophagic flux and redox systems. This explains the mechanisms by which glioma cells respond to CBD treatment. These findings highlight the therapeutic potential of CBD, suggesting that modulating NRF2 and autophagy pathways could represent a promising strategy for glioblastoma treatment.
Collapse
Affiliation(s)
- Laura Giannotti
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Benedetta Di Chiara Stanca
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), 80125 Naples, Italy
| | - Francesco Spedicato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (F.S.); (D.V.)
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy; (F.S.); (D.V.)
| | - Eleonora Stanca
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Fabrizio Damiano
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| | - Luisa Siculella
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy; (B.D.C.S.); (E.S.); (L.S.)
| |
Collapse
|
9
|
Hao X, Li Y, Gao H, Wang Z, Fang B. Inhalation Anesthetics Play a Janus-Faced Role in Self-Renewal and Differentiation of Stem Cells. Biomolecules 2024; 14:1167. [PMID: 39334933 PMCID: PMC11430341 DOI: 10.3390/biom14091167] [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: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Inhalation anesthesia stands as a pivotal modality within clinical anesthesia practices. Beyond its primary anesthetic effects, inhaled anesthetics have non-anesthetic effects, exerting bidirectional influences on the physiological state of the body and disease progression. These effects encompass impaired cognitive function, inhibition of embryonic development, influence on tumor progression, and so forth. For many years, inhaled anesthetics were viewed as inhibitors of stem cell fate regulation. However, there is now a growing appreciation that inhaled anesthetics promote stem cell biological functions and thus are now regarded as a double-edged sword affecting stem cell fate. In this review, the effects of inhaled anesthetics on self-renewal and differentiation of neural stem cells (NSCs), embryonic stem cells (ESCs), and cancer stem cells (CSCs) were summarized. The mechanisms of inhaled anesthetics involving cell cycle, metabolism, stemness, and niche of stem cells were also discussed. A comprehensive understanding of these effects will enhance our comprehension of how inhaled anesthetics impact the human body, thus promising breakthroughs in the development of novel strategies for innovative stem cell therapy approaches.
Collapse
Affiliation(s)
- Xiaotong Hao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Hairong Gao
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
10
|
Pan T, Yang B, Yao S, Wang R, Zhu Y. Exploring the multifaceted role of adenosine nucleotide translocase 2 in cellular and disease processes: A comprehensive review. Life Sci 2024; 351:122802. [PMID: 38857656 DOI: 10.1016/j.lfs.2024.122802] [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: 03/07/2024] [Revised: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Adenosine nucleotide translocases (ANTs) are a family of proteins abundant in the inner mitochondrial membrane, primarily responsible for shuttling ADP and ATP across the mitochondrial membrane. Additionally, ANTs are key players in balancing mitochondrial energy metabolism and regulating cell death. ANT2 isoform, highly expressed in undifferentiated and proliferating cells, is implicated in the development and drug resistance of various tumors. We conduct a detailed analysis of the potential mechanisms by which ANT2 may influence tumorigenesis and drug resistance. Notably, the significance of ANT2 extends beyond oncology, with roles in non-tumor cell processes including blood cell development, gastrointestinal motility, airway hydration, nonalcoholic fatty liver disease, obesity, chronic kidney disease, and myocardial development, making it a promising therapeutic target for multiple pathologies. To better understand the molecular mechanisms of ANT2, this review summarizes the structural properties, expression patterns, and basic functions of the ANT2 protein. In particular, we review and analyze the controversy surrounding ANT2, focusing on its role in transporting ADP/ATP across the inner mitochondrial membrane, its involvement in the composition of the mitochondrial permeability transition pore, and its participation in apoptosis.
Collapse
Affiliation(s)
- Tianhui Pan
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Bin Yang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Sheng Yao
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Rui Wang
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
| | - Yongliang Zhu
- Laboratory of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China.
| |
Collapse
|
11
|
Sugahara S, Brooks CR. Adenine Nucleotide Translocators Control Kidney Metabolism and Lipid Handling. J Am Soc Nephrol 2024; 35:257-258. [PMID: 38356156 PMCID: PMC10962857 DOI: 10.1681/asn.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Affiliation(s)
- Sho Sugahara
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Craig R. Brooks
- Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
12
|
Perumal N, Yurugi H, Dahm K, Rajalingam K, Grus FH, Pfeiffer N, Manicam C. Proteome landscape and interactome of voltage-gated potassium channel 1.6 (Kv1.6) of the murine ophthalmic artery and neuroretina. Int J Biol Macromol 2024; 257:128464. [PMID: 38043654 DOI: 10.1016/j.ijbiomac.2023.128464] [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: 08/29/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/05/2023]
Abstract
The voltage-gated potassium channel 1.6 (Kv1.6) plays a vital role in ocular neurovascular beds and exerts its modulatory functions via interaction with other proteins. However, the interactome and their potential roles remain unknown. Here, the global proteome landscape of the ophthalmic artery (OA) and neuroretina was mapped, followed by the determination of Kv1.6 interactome and validation of its functionality and cellular localization. Microfluorimetric analysis of intracellular [K+] and Western blot validated the native functionality and cellular expression of the recombinant Kv1.6 channel protein. A total of 54, 9 and 28 Kv1.6-interacting proteins were identified in the mouse OA and, retina of mouse and rat, respectively. The Kv1.6-protein partners in the OA, namely actin cytoplasmic 2, alpha-2-macroglobulin and apolipoprotein A-I, were implicated in the maintenance of blood vessel integrity by regulating integrin-mediated adhesion to extracellular matrix and Ca2+ flux. Many retinal protein interactors, particularly the ADP/ATP translocase 2 and cytoskeleton protein tubulin, were involved in endoplasmic reticulum stress response and cell viability. Three common interactors were found in all samples comprising heat shock cognate 71 kDa protein, Ig heavy constant gamma 1 and Kv1.6 channel. This foremost in-depth investigation enriched and identified the elusive Kv1.6 channel and, elucidated its complex interactome.
Collapse
Affiliation(s)
- Natarajan Perumal
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Hajime Yurugi
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Katrin Dahm
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Centre of the Johannes Gutenberg University Mainz, Germany
| | - Franz H Grus
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
13
|
Teraiya M, Krokhin O, Chen VC, Perreault H. Cytoplasmic Shotgun Proteomic Points to Key Proteins and Pathways in Temozolomide-Resistant Glioblastoma Multiforme. J Proteome Res 2024; 23:465-482. [PMID: 38147655 DOI: 10.1021/acs.jproteome.3c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Temozolomide (TMZ) is the first line of chemotherapy to treat primary brain tumors of the type glioblastoma multiforme (GBM). TMZ resistance (TMZR) is one of the main barriers to successful treatment and is a principal factor in relapse, resulting in a poor median survival of 15 months. The present paper focuses on proteomic analyses of cytosolic fractions from TMZ-resistant (TMZR) LN-18 cells. The experimental workflow includes an easy, cost-effective, and reproducible method to isolate subcellular fraction of cytosolic (CYTO) proteins, mitochondria, and plasma membrane proteins for proteomic studies. For this study, enriched cytoplasmic fractions were analyzed in replicates by nanoflow liquid chromatography tandem high-resolution mass spectrometry (nLC-MS/MS), and proteins identified were quantified using a label-free approach (LFQ). Statistical analysis of control (CTRL) and temozolomide-resistant (TMZR) proteomes revealed proteins that appear to be differentially controlled in the cytoplasm. The functions of these proteins are discussed as well as their roles in other cancers and TMZ resistance in GBM. Key proteins are also described through biological processes related to gene ontology (GO), molecular functions, and cellular components. For protein-protein interactions (PPI), network and pathway involvement analyses have been performed, highlighting the roles of key proteins in the TMZ resistance phenotypes. This study provides a detailed insight into methods of subcellular fractionation for proteomic analysis of TMZ-resistant GBM cells and the potential to apply this approach to future large-scale studies. Several key proteins, protein-protein interactions (PPI), and pathways have been identified, underlying the TMZ resistance phenotype and highlighting the proteins' biological functions.
Collapse
Affiliation(s)
- Milan Teraiya
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
| | - Oleg Krokhin
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
- Manitoba Centre for Proteomics and Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba R3E3P4, Canada
| | - Vincent C Chen
- Chemistry Department, Brandon University, Brandon, Manitoba R7A 6A9, Canada
| | - Hélène Perreault
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
| |
Collapse
|
14
|
Ulchenko D, Miloykovich L, Zemlyanaya O, Shimanovsky N, Fedotcheva T. Possible Participation of Adenine Nucleotide Translocase ANT1 in the Cytotoxic Action of Progestins, Glucocorticoids, and Diclofenac on Tumor Cells. Pharmaceutics 2023; 15:2787. [PMID: 38140127 PMCID: PMC10747029 DOI: 10.3390/pharmaceutics15122787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
A comparative analysis of the cytostatic effects of progestins (gestobutanoyl, megestrol acetate, amol, dienogest, and medroxyprogesterone acetate), glucocorticoids (hydrocortisone, dexamethasone), and diclofenac on tumor cells was carried out in order to confirm their in silico predicted probabilities experimentally. The results showed the different sensitivity of HeLa, MCF-7, Hep-2, K-562, and Wi-38 cell lines to progestins, glucocorticoids, and diclofenac. The minimum IC50 was found for progestin gestobutanoyl (GB) as 18 µM for HeLa cells, and varied from 31 to 38 µM for MCF-7, Hep-2, and K-562. Glucocorticoids and diclofenac were much less cytotoxic in the HeLa, MCF-7, and Hep-2 cell lines than progestins, with IC50 values in the range of 150-3000 μM. Myelogenous leukemia K-562 cells were the least sensitive to the action of progestins and glucocorticoids but the most sensitive to diclofenac, which showed a pronounced cytotoxic effect with an IC50 of 31 μM. As we have shown earlier, progestins can uniquely modulate MPTP opening via the binding of adenine nucleotide translocase. On this basis, we evaluated the expression of adenylate nucleotide translocase ANT1 (SLC25 A4) as a possible participant in cytotoxic action in these cell lines after 48 h incubation with drugs. The results showed that progestins differently regulated ANT1 expression in different cell lines. Gestobutanoyl had the opposite effect on ANT1 expression in the HeLa, K562, and Wi-38 cells compared with the other progestins. It increased the ANT1 expression more than twofold in the HeLa and K562 cells but had no influence on the Wi-38 cells. Glucocorticoids and diclofenac increased ANT1 expression in the Wi-38 cells and decreased it in the K562, MCF-7, and Hep-2 cells. The modulation of ANT1 expression discovered in our study can be a new explanation of the cytotoxic and cytoprotective effects of hormones, which can vary depending on the cell type. ANT isoforms in normal and cancerous cells could be a new target for steroid hormone and anti-inflammatory drug action.
Collapse
Affiliation(s)
| | | | | | | | - Tatiana Fedotcheva
- Science Research Laboratory of Molecular Pharmacology, Medical Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Ostrovityanova St. 1, 117997 Moscow, Russia; (D.U.); (L.M.); (O.Z.); (N.S.)
| |
Collapse
|
15
|
Solana‐Balaguer J, Martín‐Flores N, Garcia‐Segura P, Campoy‐Campos G, Pérez‐Sisqués L, Chicote‐González A, Fernández‐Irigoyen J, Santamaría E, Pérez‐Navarro E, Alberch J, Malagelada C. RTP801 mediates transneuronal toxicity in culture via extracellular vesicles. J Extracell Vesicles 2023; 12:e12378. [PMID: 37932242 PMCID: PMC10627824 DOI: 10.1002/jev2.12378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/29/2023] [Accepted: 10/17/2023] [Indexed: 11/08/2023] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication, participating in the paracrine trophic support or in the propagation of toxic molecules, including proteins. RTP801 is a stress-regulated protein, whose levels are elevated during neurodegeneration and induce neuron death. However, whether RTP801 toxicity is transferred trans-neuronally via EVs remains unknown. Hence, we overexpressed or silenced RTP801 protein in cultured cortical neurons, isolated their derived EVs (RTP801-EVs or shRTP801-EVs, respectively), and characterized EVs protein content by mass spectrometry (MS). RTP801-EVs toxicity was assessed by treating cultured neurons with these EVs and quantifying apoptotic neuron death and branching. We also tested shRTP801-EVs functionality in the pathologic in vitro model of 6-Hydroxydopamine (6-OHDA). Expression of RTP801 increased the number of EVs released by neurons. Moreover, RTP801 led to a distinct proteomic signature of neuron-derived EVs, containing more pro-apoptotic markers. Hence, we observed that RTP801-induced toxicity was transferred to neurons via EVs, activating apoptosis and impairing neuron morphology complexity. In contrast, shRTP801-EVs were able to increase the arborization in recipient neurons. The 6-OHDA neurotoxin elevated levels of RTP801 in EVs, and 6-OHDA-derived EVs lost the mTOR/Akt signalling activation via Akt and RPS6 downstream effectors. Interestingly, EVs derived from neurons where RTP801 was silenced prior to exposing them to 6-OHDA maintained Akt and RPS6 transactivation in recipient neurons. Taken together, these results suggest that RTP801-induced toxicity is transferred via EVs, and therefore, it could contribute to the progression of neurodegenerative diseases, in which RTP801 is involved.
Collapse
Affiliation(s)
- Júlia Solana‐Balaguer
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
| | - Núria Martín‐Flores
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
| | - Pol Garcia‐Segura
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
| | - Genís Campoy‐Campos
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
| | - Leticia Pérez‐Sisqués
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
| | - Almudena Chicote‐González
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
| | | | - Enrique Santamaría
- Proteored‐ISCIIIProteomics UnitNavarrabiomed, Departamento de SaludUPNAIdiSNAPamplonaSpain
| | - Esther Pérez‐Navarro
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Jordi Alberch
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Cristina Malagelada
- Department of Biomedical SciencesUniversitat de BarcelonaBarcelonaSpain
- Institut de Neurociències (UBneuro)Universitat de BarcelonaBarcelonaSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)BarcelonaSpain
| |
Collapse
|
16
|
Woo SH, Mo YJ, Lee YI, Park JH, Hwang D, Park TJ, Kang HY, Park SC, Lee YS. ANT2 Accelerates Cutaneous Wound Healing in Aged Skin by Regulating Energy Homeostasis and Inflammation. J Invest Dermatol 2023; 143:2295-2310.e17. [PMID: 37211200 DOI: 10.1016/j.jid.2023.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/23/2023]
Abstract
An effective healing response is critical to healthy aging. In particular, energy homeostasis has become increasingly recognized as a factor in effective skin regeneration. ANT2 is a mediator of adenosine triphosphate import into mitochondria for energy homeostasis. Although energy homeostasis and mitochondrial integrity are critical for wound healing, the role played by ANT2 in the repair process had not been elucidated to date. In our study, we found that ANT2 expression decreased in aged skin and cellular senescence. Interestingly, overexpression of ANT2 in aged mouse skin accelerated the healing of full-thickness cutaneous wounds. In addition, upregulation of ANT2 in replicative senescent human diploid dermal fibroblasts induced their proliferation and migration, which are critical processes in wound healing. Regarding energy homeostasis, ANT2 overexpression increased the adenosine triphosphate production rate by activating glycolysis and induced mitophagy. Notably, ANT2-mediated upregulation of HSPA6 in aged human diploid dermal fibroblasts downregulated proinflammatory genes that mediate cellular senescence and mitochondrial damage. This study shows a previously uncharacterized physiological role of ANT2 in skin wound healing by regulating cell proliferation, energy homeostasis, and inflammation. Thus, our study links energy metabolism to skin homeostasis and reports, to the best of our knowledge, a previously unreported genetic factor that enhances wound healing in an aging model.
Collapse
Affiliation(s)
- Seung-Hwa Woo
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yun Jeong Mo
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Ji Hwan Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Tae Jun Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Institution of Inflamm-aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hee Young Kang
- Institution of Inflamm-aging Translational Research Center, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Dermatology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sang Chul Park
- The Future Life & Society Research Center, Advanced Institute of Aging Science, Chonnam National University, Gwangju, Republic of Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea; Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
17
|
Zhang M, Yang Y, Zhu Z, Chen Z, Huang D. Implications of Activating the ANT2/mTOR/PGC-1α Feedback Loop: Insights into Mitochondria-Mediated Injury in Hypoxic Myocardial Cells. Curr Issues Mol Biol 2023; 45:8633-8651. [PMID: 37998720 PMCID: PMC10670450 DOI: 10.3390/cimb45110543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Mitochondrial dysfunction is known to play a critical role in the development of cardiomyocyte death during acute myocardial infarction (AMI). However, the exact mechanisms underlying this dysfunction are still under investigation. Adenine nucleotide translocase 2 (ANT2) is a key functional protein in mitochondria. We aimed at exploring the potential benefits of ANT2 inhibition against AMI. We utilized an oxygen-glucose deprivation (OGD) cell model and an AMI mice model to detect cardiomyocyte injury. We observed elevated levels of reactive oxygen species (ROS), disrupted mitochondrial membrane potential (MMP), and increased apoptosis due to the overexpression of ANT2. Additionally, we discovered that ANT2 is involved in myocardial apoptosis by activating the mTOR (mechanistic target of rapamycin kinase)-dependent PGC-1α (PPARG coactivator 1 alpha) pathway, establishing a novel feedback loop during AMI. In our experiments with AC16 cells under OGD conditions, we observed protective effects when transfected with ANT2 siRNA and miR-1203. Importantly, the overexpression of ANT2 counteracted the protective effect resulting from miR-1203 upregulation in OGD-induced AC16 cells. All these results supported that the inhibition of ANT2 could alleviate myocardial cell injury under OGD conditions. Based on these findings, we propose that RNA interference (RNAi) technology, specifically miRNA and siRNA, holds therapeutic potential by activating the ANT2/mTOR/PGC-1α feedback loop. This activation could help mitigate mitochondria-mediated injury in the context of AMI. These insights may contribute to the development of future clinical strategies for AMI.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China;
| | - Yuanzhan Yang
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Y.Y.); (Z.C.)
| | - Zhu Zhu
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China;
- Key Laboratory of Molecular Immunology, Chinese Academy of Medical Sciences, Beijing 100191, China
| | - Zixuan Chen
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Y.Y.); (Z.C.)
| | - Dongyang Huang
- Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou 515041, China;
| |
Collapse
|
18
|
Hoogstraten CA, Jacobs MME, de Boer G, van de Wal MAE, Koopman WJH, Smeitink JAM, Russel FGM, Schirris TJJ. Metabolic impact of genetic and chemical ADP/ATP carrier inhibition in renal proximal tubule epithelial cells. Arch Toxicol 2023; 97:1927-1941. [PMID: 37154957 PMCID: PMC10256673 DOI: 10.1007/s00204-023-03510-7] [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: 12/05/2022] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Mitochondrial dysfunction is pivotal in drug-induced acute kidney injury (AKI), but the underlying mechanisms remain largely unknown. Transport proteins embedded in the mitochondrial inner membrane form a significant class of potential drug off-targets. So far, most transporter-drug interactions have been reported for the mitochondrial ADP/ATP carrier (AAC). Since it remains unknown to what extent AAC contributes to drug-induced mitochondrial dysfunction in AKI, we here aimed to better understand the functional role of AAC in the energy metabolism of human renal proximal tubular cells. To this end, CRISPR/Cas9 technology was applied to generate AAC3-/- human conditionally immortalized renal proximal tubule epithelial cells. This AAC3-/- cell model was characterized with respect to mitochondrial function and morphology. To explore whether this model could provide first insights into (mitochondrial) adverse drug effects with suspicion towards AAC-mediated mechanisms, wild-type and knockout cells were exposed to established AAC inhibitors, after which cellular metabolic activity and mitochondrial respiratory capacity were measured. Two AAC3-/- clones showed a significant reduction in ADP import and ATP export rates and mitochondrial mass, without influencing overall morphology. AAC3-/- clones exhibited reduced ATP production, oxygen consumption rates and metabolic spare capacity was particularly affected, mainly in conditions with galactose as carbon source. Chemical AAC inhibition was stronger compared to genetic inhibition in AAC3-/-, suggesting functional compensation by remaining AAC isoforms in our knockout model. In conclusion, our results indicate that ciPTEC-OAT1 cells have a predominantly oxidative phenotype that was not additionally activated by switching energy source. Genetic inhibition of AAC3 particularly impacted mitochondrial spare capacity, without affecting mitochondrial morphology, suggesting an important role for AAC in maintaining the metabolic spare respiration.
Collapse
Affiliation(s)
- Charlotte A Hoogstraten
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Maaike M E Jacobs
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Guido de Boer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Melissa A E van de Wal
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Werner J H Koopman
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Department of Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| | - Jan A M Smeitink
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Khondrion BV, Nijmegen, 6525 EX, The Netherlands
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands.
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands.
| | - Tom J J Schirris
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
- Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
19
|
Sharma RK, Chafik A, Bertolin G. Aurora kinase A/AURKA functionally interacts with the mitochondrial ATP synthase to regulate energy metabolism and cell death. Cell Death Discov 2023; 9:203. [PMID: 37386025 DOI: 10.1038/s41420-023-01501-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/31/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023] Open
Abstract
Cancer cells often hijack metabolic pathways to obtain the energy required to sustain their proliferation. Understanding the molecular mechanisms underlying cancer cell metabolism is key to fine-tune the metabolic preference of specific tumors, and potentially offer new therapeutic strategies. Here, we show that the pharmacological inhibition of mitochondrial Complex V delays the cell cycle by arresting breast cancer cell models in the G0/G1 phase. Under these conditions, the abundance of the multifunctional protein Aurora kinase A/AURKA is specifically lowered. We then demonstrate that AURKA functionally interacts with the mitochondrial Complex V core subunits ATP5F1A and ATP5F1B. Altering the AURKA/ATP5F1A/ATP5F1B nexus is sufficient to trigger G0/G1 arrest, and this is accompanied by decreased glycolysis and mitochondrial respiration rates. Last, we discover that the roles of the AURKA/ATP5F1A/ATP5F1B nexus depend on the specific metabolic propensity of triple-negative breast cancer cell lines, where they correlate with cell fate. On one hand, the nexus induces G0/G1 arrest in cells relying on oxidative phosphorylation as the main source of energy. On the other hand, it allows to bypass cell cycle arrest and it triggers cell death in cells with a glycolytic metabolism. Altogether, we provide evidence that AURKA and mitochondrial Complex V subunits cooperate to maintain cell metabolism in breast cancer cells. Our work paves the way to novel anti-cancer therapies targeting the AURKA/ATP5F1A/ATP5F1B nexus to lower cancer cell metabolism and proliferation.
Collapse
Affiliation(s)
- Rakesh Kumar Sharma
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000, Rennes, France
| | - Abderrahman Chafik
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000, Rennes, France
| | - Giulia Bertolin
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, F-35000, Rennes, France.
| |
Collapse
|
20
|
Chen Y, Wu L, Liu J, Ma L, Zhang W. Adenine nucleotide translocase: Current knowledge in post-translational modifications, regulations and pathological implications for human diseases. FASEB J 2023; 37:e22953. [PMID: 37224026 DOI: 10.1096/fj.202201855rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/01/2023] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Adenine nucleotide translocases (ANTs) are central to mitochondrial integrity and bioenergetic metabolism. This review aims to integrate the progresses and knowledge on ANTs over the last few years, contributing to a potential implication of ANTs for various diseases. Structures, functions, modifications, regulators and pathological implications of ANTs for human diseases are intensively demonstrated here. ANTs have four isoforms (ANT1-4), responsible for exchanging ATP/ADP, possibly composing of pro-apoptotic mPTP as a major component, and mediating FA-dependent uncoupling of proton efflux. ANT can be modified by methylation, nitrosylation and nitroalkylation, acetylation, glutathionylation, phosphorylation, carbonylation and hydroxynonenal-induced modifications. Compounds, including bongkrekic acid, atractyloside calcium, carbon monoxide, minocycline, 4-(N-(S-penicillaminylacetyl)amino) phenylarsonous acid, cardiolipin, free long-chain fatty acids, agaric acid, long chain acyl-coenzyme A esters, all have an ability to regulate ANT activities. ANT impairment leads to bioenergetic failure and mitochondrial dysfunction, contributing to pathogenesis of diseases, such as diabetes (deficiency), heart disease (deficiency), Parkinson's disease (reduction), Sengers Syndrome (decrease), cancer (isoform shifting), Alzheimer's Disease (coaggregation with Tau), Progressive External Opthalmoplegia (mutation), and Fascioscapulohumeral muscular dystrophy (overexpression). This review improves the understanding of the mechanism of ANT in pathogenesis of human diseases, and opens a window for novel therapeutic strategies targeted on ANT in diseases.
Collapse
Affiliation(s)
- Yingfei Chen
- Grade 2020, Capital Medical University, Beijing, China
| | - Leshuang Wu
- Grade 2019, Dalian Medical University, Dalian, China
| | - Jun Liu
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Li Ma
- Department of Epidemiology, Dalian Medical University, Dalian, China
| | - Wenli Zhang
- Biochemistry and Molecular Biology Department of College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
21
|
Seneviratne JA, Carter DR, Mittra R, Gifford A, Kim PY, Luo J, Mayoh C, Salib A, Rahmanto AS, Murray J, Cheng NC, Nagy Z, Wang Q, Kleynhans A, Tan O, Sutton SK, Xue C, Chung SA, Zhang Y, Sun C, Zhang L, Haber M, Norris MD, Fletcher JI, Liu T, Dilda PJ, Hogg PJ, Cheung BB, Marshall GM. Inhibition of mitochondrial translocase SLC25A5 and histone deacetylation is an effective combination therapy in neuroblastoma. Int J Cancer 2023; 152:1399-1413. [PMID: 36346110 PMCID: PMC10953412 DOI: 10.1002/ijc.34349] [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: 02/16/2022] [Revised: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
The mitochondrion is a gatekeeper of apoptotic processes, and mediates drug resistance to several chemotherapy agents used to treat cancer. Neuroblastoma is a common solid cancer in young children with poor clinical outcomes following conventional chemotherapy. We sought druggable mitochondrial protein targets in neuroblastoma cells. Among mitochondria-associated gene targets, we found that high expression of the mitochondrial adenine nucleotide translocase 2 (SLC25A5/ANT2), was a strong predictor of poor neuroblastoma patient prognosis and contributed to a more malignant phenotype in pre-clinical models. Inhibiting this transporter with PENAO reduced cell viability in a panel of neuroblastoma cell lines in a TP53-status-dependant manner. We identified the histone deacetylase inhibitor, suberanilohydroxamic acid (SAHA), as the most effective drug in clinical use against mutant TP53 neuroblastoma cells. SAHA and PENAO synergistically reduced cell viability, and induced apoptosis, in neuroblastoma cells independent of TP53-status. The SAHA and PENAO drug combination significantly delayed tumour progression in pre-clinical neuroblastoma mouse models, suggesting that these clinically advanced inhibitors may be effective in treating the disease.
Collapse
Affiliation(s)
- Janith A. Seneviratne
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Daniel R. Carter
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- School of Biomedical EngineeringUniversity of Technology SydneyNew South WalesAustralia
| | - Rituparna Mittra
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Andrew Gifford
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalRandwickNew South WalesAustralia
| | - Patrick Y. Kim
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Jie‐Si Luo
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Chelsea Mayoh
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Alice Salib
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Aldwin S. Rahmanto
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Jayne Murray
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Ngan C. Cheng
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Zsuzsanna Nagy
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Qian Wang
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Ane Kleynhans
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Owen Tan
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Selina K. Sutton
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Chengyuan Xue
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Sylvia A. Chung
- Adult Cancer Program, Lowy Cancer Research CentreUNSW SydneyNew South WalesAustralia
| | - Yizhuo Zhang
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Chengtao Sun
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Li Zhang
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Murray D. Norris
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- University of New South WalesCentre for Childhood Cancer ResearchRandwickNew South WalesAustralia
| | - Jamie I. Fletcher
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Pierre J. Dilda
- Adult Cancer Program, Lowy Cancer Research CentreUNSW SydneyNew South WalesAustralia
| | - Philip J. Hogg
- Australian Cancer Research Foundation (ACRF), Centenary Cancer Research Centre, Charles Perkins CentreUniversity of SydneyNew South WalesAustralia
| | - Belamy B. Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Glenn M. Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalRandwickNew South WalesAustralia
| |
Collapse
|
22
|
Chen W, Chiang J, Shang Z, Palchik G, Newman C, Zhang Y, Davis AJ, Lee H, Chen BPC. DNA-PKcs and ATM modulate mitochondrial ADP-ATP exchange as an oxidative stress checkpoint mechanism. EMBO J 2023; 42:e112094. [PMID: 36727301 PMCID: PMC10015379 DOI: 10.15252/embj.2022112094] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/13/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023] Open
Abstract
DNA-PKcs is a key regulator of DNA double-strand break repair. Apart from its canonical role in the DNA damage response, DNA-PKcs is involved in the cellular response to oxidative stress (OS), but its exact role remains unclear. Here, we report that DNA-PKcs-deficient human cells display depolarized mitochondria membrane potential (MMP) and reoriented metabolism, supporting a role for DNA-PKcs in oxidative phosphorylation (OXPHOS). DNA-PKcs directly interacts with mitochondria proteins ANT2 and VDAC2, and formation of the DNA-PKcs/ANT2/VDAC2 (DAV) complex supports optimal exchange of ADP and ATP across mitochondrial membranes to energize the cell via OXPHOS and to maintain MMP. Moreover, we demonstrate that the DAV complex temporarily dissociates in response to oxidative stress to attenuate ADP-ATP exchange, a rate-limiting step for OXPHOS. Finally, we found that dissociation of the DAV complex is mediated by phosphorylation of DNA-PKcs at its Thr2609 cluster by ATM kinase. Based on these findings, we propose that the coordination between the DAV complex and ATM serves as a novel oxidative stress checkpoint to decrease ROS production from mitochondrial OXPHOS and to hasten cellular recovery from OS.
Collapse
Affiliation(s)
- Wei‐Min Chen
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Jui‐Chung Chiang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Zengfu Shang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Guillermo Palchik
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Ciara Newman
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Yuanyuan Zhang
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Anthony J Davis
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| | - Hsinyu Lee
- Department of Life ScienceNational Taiwan UniversityTaipeiTaiwan
| | - Benjamin PC Chen
- Division of Molecular Radiation Biology, Department of Radiation OncologyUniversity of Texas Southwestern Medical Center at DallasDallasTXUSA
| |
Collapse
|
23
|
Waseem M, Wang BD. Promising Strategy of mPTP Modulation in Cancer Therapy: An Emerging Progress and Future Insight. Int J Mol Sci 2023; 24:5564. [PMID: 36982637 PMCID: PMC10051994 DOI: 10.3390/ijms24065564] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Cancer has been progressively a major global health concern. With this developing global concern, cancer determent is one of the most significant public health challenges of this era. To date, the scientific community undoubtedly highlights mitochondrial dysfunction as a hallmark of cancer cells. Permeabilization of the mitochondrial membranes has been implicated as the most considerable footprint in apoptosis-mediated cancer cell death. Under the condition of mitochondrial calcium overload, exclusively mediated by oxidative stress, an opening of a nonspecific channel with a well-defined diameter in mitochondrial membrane allows free exchange between the mitochondrial matrix and the extra mitochondrial cytosol of solutes and proteins up to 1.5 kDa. Such a channel/nonspecific pore is recognized as the mitochondrial permeability transition pore (mPTP). mPTP has been established for regulating apoptosis-mediated cancer cell death. It has been evident that mPTP is critically linked with the glycolytic enzyme hexokinase II to defend cellular death and reduce cytochrome c release. However, elevated mitochondrial Ca2+ loading, oxidative stress, and mitochondrial depolarization are critical factors leading to mPTP opening/activation. Although the exact mechanism underlying mPTP-mediated cell death remains elusive, mPTP-mediated apoptosis machinery has been considered as an important clamp and plays a critical role in the pathogenesis of several types of cancers. In this review, we focus on structure and regulation of the mPTP complex-mediated apoptosis mechanisms and follow with a comprehensive discussion addressing the development of novel mPTP-targeting drugs/molecules in cancer treatment.
Collapse
Affiliation(s)
- Mohammad Waseem
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA;
- Hormone Related Cancers Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| |
Collapse
|
24
|
Preservation of mitochondrial membrane potential is necessary for lifespan extension from dietary restriction. GeroScience 2023:10.1007/s11357-023-00766-w. [PMID: 36877298 PMCID: PMC10400507 DOI: 10.1007/s11357-023-00766-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Dietary restriction (DR) increases lifespan in many organisms, but its underlying mechanisms are not fully understood. Mitochondria play a central role in metabolic regulation and are known to undergo changes in structure and function in response to DR. Mitochondrial membrane potential (Δψm) is the driving force for ATP production and mitochondrial outputs that integrate many cellular signals. One such signal regulated by Δψm is nutrient-status sensing. Here, we tested the hypothesis that DR promotes longevity through preserved Δψm during adulthood. Using the nematode Caenorhabditis elegans, we find that Δψm declines with age relatively early in the lifespan, and this decline is attenuated by DR. Pharmacologic depletion of Δψm blocked the longevity and health benefits of DR. Genetic perturbation of Δψm and mitochondrial ATP availability similarly prevented lifespan extension from DR. Taken together, this study provides further evidence that appropriate regulation of Δψm is a critical factor for health and longevity in response to DR.
Collapse
|
25
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
26
|
Shu Y, Hao Y, Feng J, Liu H, Li S, Feng J, Jiang Z, Ye L, Zhou Y, Sun Y, Zhou Z, Wei H, Gao P, Zhang H, Sun L. Non-canonical phosphoglycerate dehydrogenase activity promotes liver cancer growth via mitochondrial translation and respiratory metabolism. EMBO J 2022; 41:e111550. [PMID: 36314841 PMCID: PMC9713714 DOI: 10.15252/embj.2022111550] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Phosphoglycerate dehydrogenase (PHGDH) is a key serine biosynthesis enzyme whose aberrant expression promotes various types of tumors. Recently, PHGDH has been found to have some non-canonical functions beyond serine biosynthesis, but its specific mechanisms in tumorigenesis remain unclear. Here, we show that PHGDH localizes to the inner mitochondrial membrane and promotes the translation of mitochondrial DNA (mtDNA)-encoded proteins in liver cancer cells. Mechanistically, we demonstrate that mitochondrial PHGDH directly interacts with adenine nucleotide translocase 2 (ANT2) and then recruits mitochondrial elongation factor G2 (mtEFG2) to promote mitochondrial ribosome recycling efficiency, thereby promoting mtDNA-encoded protein expression and subsequent mitochondrial respiration. Moreover, we show that treatment with a mitochondrial translation inhibitor or depletion of mtEFG2 diminishes PHGDH-mediated tumor growth. Collectively, our findings uncover a previously unappreciated function of PHGDH in tumorigenesis acting via promotion of mitochondrial translation and bioenergetics.
Collapse
Affiliation(s)
- Ying Shu
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yijie Hao
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Junru Feng
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Haiying Liu
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Shi‐ting Li
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Jiaqian Feng
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Zetan Jiang
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Ling Ye
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Yingli Zhou
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Yuchen Sun
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Zilong Zhou
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Haoran Wei
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Ping Gao
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Huafeng Zhang
- Division of Life Science and Medicine, Anhui Key Laboratory of Hepatopancreatobiliary Surgery, Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, The Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical SciencesUniversity of Science and Technology of ChinaHefeiChina
- Division of Life Science and Medicine, Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Linchong Sun
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| |
Collapse
|
27
|
Tonner H, Hunn S, Auler N, Schmelter C, Pfeiffer N, Grus FH. Dynamin-like Protein 1 (DNML1) as a Molecular Target for Antibody-Based Immunotherapy to Treat Glaucoma. Int J Mol Sci 2022; 23:ijms232113618. [PMID: 36362420 PMCID: PMC9654827 DOI: 10.3390/ijms232113618] [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: 09/01/2022] [Revised: 09/23/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
Slow and progressive loss of retinal ganglion cells (RGCs) is the main characteristic of glaucoma, the second leading cause of blindness worldwide. Previous studies have shown that impaired mitochondrial dynamics could facilitate retinal neurodegeneration. Mitochondrial dynamics are regulated directly (fission) or more indirectly (fusion) by dynamin-like protein 1 (DNML1). Therefore, DNM1L might be a promising target for an antibody-based approach to treat glaucoma. The consequences of targeting endogenous DNM1L by antibodies in a glaucoma animal model have not been investigated yet. Here, we show that the intravitreal application of an anti-DNM1L antibody showed protective effects regarding the survival of RGCs and their axons in the retinal nerve fiber layer (RNFL). Antibody treatment also improved retinal functionality, as observed by electroretinography (Ganzfeld ERG). Western blot analysis revealed altered DNM1L phosphorylation and altered expression of proteins related to apoptosis suggesting a decreased apoptosis rate. Mass spectrometry analysis revealed 28 up-regulated and 21 down-regulated proteins (p < 0.05) in both experimental groups. Protein pathway analysis showed that many proteins interacted directly with the target protein DNM1L and could be classified into three main protein clusters: Vesicle traffic-associated (NSF, SNCA, ARF1), mitochondrion-associated (HSP9A, SLC25A5/ANT2, GLUD1) and cytoskeleton-associated (MAP1A) signaling pathway. Our results demonstrate that DNM1L is a promising target for an antibody-based approach to glaucoma therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Franz H. Grus
- Correspondence: ; Tel.: +49-6131-17-3328; Fax: +49-6131-4970563
| |
Collapse
|
28
|
Liu AR, Liu YN, Shen SX, Yan LR, Lv Z, Ding HX, Wang A, Yuan Y, Xu Q. Comprehensive Analysis and Validation of Solute Carrier Family 25 (SLC25) and Its Correlation with Immune Infiltration in Pan-Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4009354. [PMID: 36254139 PMCID: PMC9569204 DOI: 10.1155/2022/4009354] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022]
Abstract
As the largest gene family functioning in protein transport among human solute carriers, the SLC25 family (mitochondrial carrier family) can participate in development of cancer. However, a comprehensive exploration for the exactly roles of SLC family remains lacking. In the present study, a total of 15 functional SLC25 family genes were retrieved from all current publications. And multidimensional analyses were systematically performed based on the transcriptome and genome data of SLC25 family from a variety of online databases for their expression, immune cell infiltration, and cancer prognosis. Validation by qPCR and immunohistochemistry were further conducted for the expression of partial SLC25 family members in some tumor tissue. We found that the SLC25 family had strong correlation with immune cells, such as macrophages M2, CD8+ T cell, CD4+ T cell memory activated, and memory resting. Among them, SLC25A6 was most correlated with Macrophage M1 in uveal melanoma (r = -0.68, P = 1.9e - 0.5). Expression of mRNA level showed that SLC25A4 was downregulated in stomach adenocarcinoma and colon adenocarcinoma. SLC25A7 was highly expressed in stomach adenocarcinoma and colon adenocarcinoma. SLC25A23 was decreased in colon adenocarcinoma. qPCR and immunohistochemistry validation results were consistent with our bioinformatics prediction. SLC25A8 was associated with the prognosis of cancer. All these findings suggested that the SLC25 family might affects the immune microenvironment of the cancer and then had the potential to be predictive biomarkers for early diagnosis and prognosis as well as novel targets for individualized treatment of cancer.
Collapse
Affiliation(s)
- Ao-ran Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ying-nan Liu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shi-xuan Shen
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Li-rong Yan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhi Lv
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Han-xi Ding
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ang Wang
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yuan Yuan
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| | - Qian Xu
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Hospital of China Medical University, No. 155 North NanjingBei Street, Heping District, Shenyang, 110001 Liaoning, China
- Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, China
- Key Laboratory of GI Cancer Etiology and Prevention in Liaoning Province, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
29
|
Gottschalk B, Koshenov Z, Bachkoenig OA, Rost R, Malli R, Graier WF. MFN2 mediates ER-mitochondrial coupling during ER stress through specialized stable contact sites. Front Cell Dev Biol 2022; 10:918691. [PMID: 36158213 PMCID: PMC9493370 DOI: 10.3389/fcell.2022.918691] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Endoplasmic reticulum (ER) functions critically depend on a suitable ATP supply to fuel ER chaperons and protein trafficking. A disruption of the ability of the ER to traffic and fold proteins leads to ER stress and the unfolded protein response (UPR). Using structured illumination super-resolution microscopy, we revealed increased stability and lifetime of mitochondrial associated ER membranes (MAM) during ER stress. The consequent increase of basal mitochondrial Ca2+ leads to increased TCA cycle activity and enhanced mitochondrial membrane potential, OXPHOS, and ATP generation during ER stress. Subsequently, OXPHOS derived ATP trafficking towards the ER was increased. We found that the increased lifetime and stability of MAMs during ER stress depended on the mitochondrial fusion protein Mitofusin2 (MFN2). Knockdown of MFN2 blunted mitochondrial Ca2+ effect during ER stress, switched mitochondrial F1FO-ATPase activity into reverse mode, and strongly reduced the ATP supply for the ER during ER stress. These findings suggest a critical role of MFN2-dependent MAM stability and lifetime during ER stress to compensate UPR by strengthening ER ATP supply by the mitochondria.
Collapse
Affiliation(s)
- Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - René Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
30
|
Barnstable CJ, Zhang M, Tombran-Tink J. Uncoupling Proteins as Therapeutic Targets for Neurodegenerative Diseases. Int J Mol Sci 2022; 23:5672. [PMID: 35628482 PMCID: PMC9144266 DOI: 10.3390/ijms23105672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 02/05/2023] Open
Abstract
Most of the major retinal degenerative diseases are associated with significant levels of oxidative stress. One of the major sources contributing to the overall level of stress is the reactive oxygen species (ROS) generated by mitochondria. The driving force for ROS production is the proton gradient across the inner mitochondrial membrane. This gradient can be modulated by members of the uncoupling protein family, particularly the widely expressed UCP2. The overexpression and knockout studies of UCP2 in mice have established the ability of this protein to provide neuroprotection in a number of animal models of neurological disease, including retinal diseases. The expression and activity of UCP2 are controlled at the transcriptional, translational and post-translational levels, making it an ideal candidate for therapeutic intervention. In addition to regulation by a number of growth factors, including the neuroprotective factors LIF and PEDF, small molecule activators of UCP2 have been found to reduce mitochondrial ROS production and protect against cell death both in culture and animal models of retinal degeneration. Such studies point to the development of new therapeutics to combat a range of blinding retinal degenerative diseases and possibly other diseases in which oxidative stress plays a key role.
Collapse
Affiliation(s)
- Colin J. Barnstable
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA;
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin 300384, China;
| | - Mingliang Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin 300384, China;
| | - Joyce Tombran-Tink
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA;
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, 251 Fukang Road, Tianjin 300384, China;
| |
Collapse
|
31
|
Adamia S, Bhatt S, Wen K, Chyra Z, Fell GG, Tai YT, Pioso MS, Abiatari I, Letai A, Dorfman DM, Hideshima T, Anderson KC. Combination therapy targeting Erk1/2 and CDK4/6i in relapsed refractory multiple myeloma. Leukemia 2022; 36:1088-1101. [PMID: 35082402 PMCID: PMC8979823 DOI: 10.1038/s41375-021-01475-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 11/09/2022]
Abstract
Oncogenic activated RAS mutations have been detected in 50% of de novo and 70% of relapsed multiple myeloma (MM) patients. Translocation t(11;14) involving IgH/CCDN1 and overexpression of cyclin-Ds are early events in MM pathogenesis, enhancing uncontrolled MM cell growth. We hypothesized that targeting both RAS/MAPK pathway molecules including Erk1/2 along with cyclin-Ds enhances MM cytotoxicity and minimizes side effects. Recent studies have demonstrated the high potency of Erk1/2 and CDK4/6 inhibitors in metastatic relapsed cancers, and here we tested anti-MM effects of the Erk1/2 + CDK4/6 inhibitor combination. Our studies showed strong synergistic (IC < 0.5) cytotoxicity of Erk1/2i + CDK4/6i in MM-cells. Erk1/2i + CDK4/6i treatment in a dose-dependent manner arrested MM-cells in the G0/G1 phase and activated mitochondrial apoptotic signaling. Our studies showed that Erk1/2i + CDK4/6i treatment-induced inhibition of key target molecules in Erk1/2 and CDK4/6 signaling, such as c-myc, p-RSK, p-S6, p-RB, and E2F1, suggesting on-target activity of these inhibitors. We identified Erk1/2i + CDK4/6i treatment associated five-gene signature which includes SNRPB and SLC25A5; these genes are involved in RNA processing and mitochondrial metabolism, respectively. Overall, our studies provide the preclinical framework for Erk1/2i + CDK4/6i combination clinical trials to target Ras+CDK pathways to improve patient outcome in MM.
Collapse
Affiliation(s)
- Sophia Adamia
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
| | - Shruti Bhatt
- Dana-FArber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.,Department of Pharmacy, National University of Singapore, Singapore, 117559, Singapore
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Zuzana Chyra
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Geoffrey G Fell
- Dana-Farber Cancer Institute, Department of Data science, Boston, MA, 02215, USA
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Marisa S Pioso
- Dana-FArber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Ivane Abiatari
- Ilia State University, School of Medicine, Tbilisi, G409, Georgia
| | - Anthony Letai
- Dana-FArber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02215, USA
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
32
|
Saccharomyces cerevisiae as a Tool for Studying Mutations in Nuclear Genes Involved in Diseases Caused by Mitochondrial DNA Instability. Genes (Basel) 2021; 12:genes12121866. [PMID: 34946817 PMCID: PMC8701800 DOI: 10.3390/genes12121866] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023] Open
Abstract
Mitochondrial DNA (mtDNA) maintenance is critical for oxidative phosphorylation (OXPHOS) since some subunits of the respiratory chain complexes are mitochondrially encoded. Pathological mutations in nuclear genes involved in the mtDNA metabolism may result in a quantitative decrease in mtDNA levels, referred to as mtDNA depletion, or in qualitative defects in mtDNA, especially in multiple deletions. Since, in the last decade, most of the novel mutations have been identified through whole-exome sequencing, it is crucial to confirm the pathogenicity by functional analysis in the appropriate model systems. Among these, the yeast Saccharomyces cerevisiae has proved to be a good model for studying mutations associated with mtDNA instability. This review focuses on the use of yeast for evaluating the pathogenicity of mutations in six genes, MPV17/SYM1, MRM2/MRM2, OPA1/MGM1, POLG/MIP1, RRM2B/RNR2, and SLC25A4/AAC2, all associated with mtDNA depletion or multiple deletions. We highlight the techniques used to construct a specific model and to measure the mtDNA instability as well as the main results obtained. We then report the contribution that yeast has given in understanding the pathogenic mechanisms of the mutant variants, in finding the genetic suppressors of the mitochondrial defects and in the discovery of molecules able to improve the mtDNA stability.
Collapse
|
33
|
Zhang H, Chen N, Deng Z, Mai Y, Deng L, Chen G, Li Y, Pan B, Zhong W. Suppression of ANT2 by miR-137 Inhibits Prostate Tumorigenesis. Front Genet 2021; 12:687236. [PMID: 34539732 PMCID: PMC8448070 DOI: 10.3389/fgene.2021.687236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is a serious disease that affects men’s health. To date, no effective and long-lasting treatment option for this condition is available in clinical practice. ANT2 is highly expressed in a variety of hormone-related cancers, but its relationship and regulatory mechanism with PCa are unclear. In this study, we found that ANT2 expression was significantly upregulated in PCa tissues relative to control samples. Genetic knockdown of ANT2 effectively inhibited, while overexpression promoted, proliferation, migration, and invasion of PCa cells. In addition, miR-137 expression was reduced in prostate cancer tissues relative to control tissues. We identified a regulatory site for miR-137 in the 3′-UTR of ANT2 mRNA; luciferase reporter assays indicated that ANT2 is a direct target gene for miR-137. Transfecting cells with miR-137 mimics and/or an ANT2-encoding plasmid revealed that ANT2 promotes proliferation, migration, and invasion of PCa, whereas co-expression of miR-137 mimics inhibited these behaviors. These observations suggest that miR-137 mimics inhibit development of PCa by antagonizing expression of ANT2. Furthermore, tumorigenic assays in nude mice showed that miR-137 inhibitors abolished the inhibitory effect of ANT2 knockdown on PCa tumor growth. Collectively, our findings suggest that ANT2, a target gene of miR-137, is intimately involved in development of PCa, providing new evidence for the mechanism underlying pathogenesis of PCa as well as new options for targeted therapy.
Collapse
Affiliation(s)
- Heyuan Zhang
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Nanhui Chen
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Zhihai Deng
- Department of Urology, Gaozhou People's Hospital, Gaozhou, China
| | - Yang Mai
- Department of Urology, Guangzhou Twelfth People's Hospital, Guangzhou, China
| | - Limin Deng
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yutong Li
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Pan
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weifeng Zhong
- Department of Urology, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China.,Department of Urology, Guangzhou Twelfth People's Hospital, Guangzhou, China
| |
Collapse
|
34
|
Mitochondrial Disruption by Amyloid Beta 42 Identified by Proteomics and Pathway Mapping. Cells 2021; 10:cells10092380. [PMID: 34572029 PMCID: PMC8468661 DOI: 10.3390/cells10092380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is marked by chronic neurodegeneration associated with the occurrence of plaques containing amyloid β (Aβ) proteins in various parts of the human brain. An increase in several Aβ fragments is well documented in patients with AD and anti-amyloid targeting is an emerging area of therapy. Soluble Aβ can bind to various cell surface and intracellular molecules with the pathogenic Aβ42 fragment leading to neurotoxicity. Here we examined the effect of Aβ42 on network adaptations in the proteome of nerve growth factor (NGF) differentiated PC12 cells using liquid-chromatography electrospray ionization mass spectrometry (LC-ESI MS/MS) proteomics. Whole-cell peptide mass fingerprinting was coupled to bioinformatic gene set enrichment analysis (GSEA) in order to identify differentially represented proteins and related gene ontology (GO) pathways within Aβ42 treated cells. Our results underscore a role for Aβ42 in disrupting proteome responses for signaling, bioenergetics, and morphology in mitochondria. These findings highlight the specific components of the mitochondrial response during Aβ42 neurotoxicity and suggest several new biomarkers for detection and surveillance of amyloid disease.
Collapse
|
35
|
Ferramosca A, Zara V. Mitochondrial Carriers and Substrates Transport Network: A Lesson from Saccharomyces cerevisiae. Int J Mol Sci 2021; 22:ijms22168496. [PMID: 34445202 PMCID: PMC8395155 DOI: 10.3390/ijms22168496] [Citation(s) in RCA: 12] [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: 07/20/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
The yeast Saccharomyces cerevisiae is one of the most widely used model organisms for investigating various aspects of basic cellular functions that are conserved in human cells. This organism, as well as human cells, can modulate its metabolism in response to specific growth conditions, different environmental changes, and nutrient depletion. This adaptation results in a metabolic reprogramming of specific metabolic pathways. Mitochondrial carriers play a fundamental role in cellular metabolism, connecting mitochondrial with cytosolic reactions. By transporting substrates across the inner membrane of mitochondria, they contribute to many processes that are central to cellular function. The genome of Saccharomyces cerevisiae encodes 35 members of the mitochondrial carrier family, most of which have been functionally characterized. The aim of this review is to describe the role of the so far identified yeast mitochondrial carriers in cell metabolism, attempting to show the functional connections between substrates transport and specific metabolic pathways, such as oxidative phosphorylation, lipid metabolism, gluconeogenesis, and amino acids synthesis. Analysis of the literature reveals that these proteins transport substrates involved in the same metabolic pathway with a high degree of flexibility and coordination. The understanding of the role of mitochondrial carriers in yeast biology and metabolism could be useful for clarifying unexplored aspects related to the mitochondrial carrier network. Such knowledge will hopefully help in obtaining more insight into the molecular basis of human diseases.
Collapse
|
36
|
Li S, Liu S, Dai Z, Zhang Q, Xu Y, Chen Y, Jiang Z, Huang W, Sun H. The UL16 protein of HSV-1 promotes the metabolism of cell mitochondria by binding to ANT2 protein. Sci Rep 2021; 11:14001. [PMID: 34234233 PMCID: PMC8263751 DOI: 10.1038/s41598-021-93430-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/15/2021] [Indexed: 11/09/2022] Open
Abstract
Long-term studies have shown that virus infection affects the energy metabolism of host cells, which mainly affects the function of mitochondria and leads to the hydrolysis of ATP in host cells, but it is not clear how virus infection participates in mitochondrial energy metabolism in host cells. In our study, HUVEC cells were infected with HSV-1, and the differentially expressed genes were obtained by microarray analysis and data analysis. The viral gene encoding protein UL16 was identified to interact with host protein ANT2 by immunoprecipitation and mass spectrometry. We also reported that UL16 transfection promoted oxidative phosphorylation of glucose and significantly increased intracellular ATP content. Furthermore, UL16 was transfected into the HUVEC cell model with mitochondrial dysfunction induced by d-Gal, and it was found that UL16 could restore the mitochondrial function of cells. It was first discovered that viral protein UL16 could enhance mitochondrial function in mammalian cells by promoting mitochondrial metabolism. This study provides a theoretical basis for the prevention and treatment of mitochondrial dysfunction or the pathological process related to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Shiyu Li
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, China.,Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuting Liu
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhenning Dai
- Department of Stomatology, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou, 510095, China
| | - Qian Zhang
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yichao Xu
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Youyu Chen
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Zhenyou Jiang
- Department of Microbiology and Immunology, College of Basic Medicine and Public Hygiene, Jinan University, Guangzhou, 510632, China.
| | - Wenhua Huang
- Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Hanxiao Sun
- Institute of Genomic Medicine, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
37
|
Lemasters JJ. Metabolic implications of non-electrogenic ATP/ADP exchange in cancer cells: A mechanistic basis for the Warburg effect. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2021; 1862:148410. [PMID: 33722515 PMCID: PMC8096716 DOI: 10.1016/j.bbabio.2021.148410] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/07/2021] [Indexed: 12/20/2022]
Abstract
In post-mitotic cells, mitochondrial ATP/ADP exchange occurs by the adenine nucleotide translocator (ANT). Driven by membrane potential (ΔΨ), ANT catalyzes electrogenic exchange of ATP4- for ADP3-, leading to higher ATP/ADP ratios in the cytosol than mitochondria. In cancer cells, ATP/ADP exchange occurs not by ANT but likely via the non-electrogenic ATP-Mg/phosphate carrier. Consequences of non-electrogenic exchange are: 1) Cytosolic ATP/ADP decreases to stimulate aerobic glycolysis. 2) Without proton utilization for exchange, ATP/O increases by 35% for complete glucose oxidation. 3) Decreased cytosolic ATP/ADPPi increases NAD(P)H/NAD(P)+. Increased NADH increases lactate/pyruvate, and increased NADPH promotes anabolic metabolism. Fourth, increased mitochondrial NADH/NAD+ magnifies the redox span across Complexes I and III, which increases ΔΨ, reactive oxygen species generation, and susceptibility to ferroptosis. 5) Increased mitochondrial NADPH/NADP+ favors a reverse isocitrate dehydrogenase-2 reaction with citrate accumulation and export for biomass formation. Consequently, 2-oxoglutarate formation occurs largely via oxidation of glutamine, the preferred respiratory substrate of cancer cells. Overall, non-electrogenic ATP/ADP exchange promotes aerobic glycolysis (Warburg effect) and confers specific growth advantages to cancer cells.
Collapse
Affiliation(s)
- John J Lemasters
- Center for Cell Death, Injury & Regeneration, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States of America; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States of America.
| |
Collapse
|
38
|
Hiu JJ, Yap MKK. The effects of Naja sumatrana venom cytotoxin, sumaCTX on alteration of the secretome in MCF-7 breast cancer cells following membrane permeabilization. Int J Biol Macromol 2021; 184:776-786. [PMID: 34174307 DOI: 10.1016/j.ijbiomac.2021.06.145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022]
Abstract
Naja sumatrana venom cytotoxin (sumaCTX) is a basic protein which belongs to three-finger toxin family. It has been shown to induce caspase-dependent, mitochondrial-mediated apoptosis in MCF-7 cells at lower concentrations. This study aimed to investigate the alteration of secretome in MCF-7 cells following membrane permeabilization by high concentrations of sumaCTX, using label-free quantitative (LFQ) approach. The degree of membrane permeabilization of sumaCTX was determined by lactate dehydrogenase (LDH) assay and calcein-propidium iodide (PI) assays. LDH and calcein-PI assays revealed time-dependent membrane permeabilization within a narrow concentration range. However, as toxin concentrations increased, prolonged exposure of MCF-7 cells to sumaCTX did not promote the progression of membrane permeabilization. The secretome analyses showed that membrane permeabilization was an event preceding the release of intracellular proteins. Bioinformatics analyses of the LFQ secretome revealed the presence of 105 significantly distinguished proteins involved in metabolism, structural supports, inflammatory responses, and necroptosis in MCF-7 cells treated with 29.8 μg/mL of sumaCTX. Necroptosis was presumably an initial stress response in MCF-7 cells when exposed to high sumaCTX concentration. Collectively, sumaCTX-induced the loss of membrane integrity in a concentration-dependent manner, whereby the cell death pattern of MCF-7 cells transformed from apoptosis to necroptosis with increasing toxin concentrations.
Collapse
Affiliation(s)
- Jia Jin Hiu
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia.
| | - Michelle Khai Khun Yap
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia; Tropical Medicine and Biology Multidisciplinary Platform, Monash University Malaysia, Bandar Sunway, Malaysia.
| |
Collapse
|
39
|
Nelson MAM, McLaughlin KL, Hagen JT, Coalson HS, Schmidt C, Kassai M, Kew KA, McClung JM, Neufer PD, Brophy P, Vohra NA, Liles D, Cabot MC, Fisher-Wellman KH. Intrinsic OXPHOS limitations underlie cellular bioenergetics in leukemia. eLife 2021; 10:e63104. [PMID: 34132194 PMCID: PMC8221809 DOI: 10.7554/elife.63104] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
Currently there is great interest in targeting mitochondrial oxidative phosphorylation (OXPHOS) in cancer. However, notwithstanding the targeting of mutant dehydrogenases, nearly all hopeful 'mito-therapeutics' cannot discriminate cancerous from non-cancerous OXPHOS and thus suffer from a limited therapeutic index. Using acute myeloid leukemia (AML) as a model, herein, we leveraged an in-house diagnostic biochemical workflow to identify 'actionable' bioenergetic vulnerabilities intrinsic to cancerous mitochondria. Consistent with prior reports, AML growth and proliferation was associated with a hyper-metabolic phenotype which included increases in basal and maximal respiration. However, despite having nearly 2-fold more mitochondria per cell, clonally expanding hematopoietic stem cells, leukemic blasts, as well as chemoresistant AML were all consistently hallmarked by intrinsic OXPHOS limitations. Remarkably, by performing experiments across a physiological span of ATP free energy, we provide direct evidence that leukemic mitochondria are particularly poised to consume ATP. Relevant to AML biology, acute restoration of oxidative ATP synthesis proved highly cytotoxic to leukemic blasts, suggesting that active OXPHOS repression supports aggressive disease dissemination in AML. Together, these findings argue against ATP being the primary output of leukemic mitochondria and provide proof-of-principle that restoring, rather than disrupting, OXPHOS may represent an untapped therapeutic avenue for combatting hematological malignancy and chemoresistance.
Collapse
Affiliation(s)
- Margaret AM Nelson
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Kelsey L McLaughlin
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - James T Hagen
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Hannah S Coalson
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Cameron Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Miki Kassai
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - Kimberly A Kew
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - Joseph M McClung
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
- Department of Cardiovascular Sciences, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Patricia Brophy
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| | - Nasreen A Vohra
- Department of Surgery, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - Darla Liles
- Department of Internal Medicine, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - Myles C Cabot
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
| | - Kelsey H Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina UniversityGreenvilleUnited States
- East Carolina Diabetes and Obesity Institute, East Carolina UniversityGreenvilleUnited States
| |
Collapse
|
40
|
Ambekar T, Pawar J, Rathod R, Patel M, Fernandes V, Kumar R, Singh SB, Khatri DK. Mitochondrial quality control: Epigenetic signatures and therapeutic strategies. Neurochem Int 2021; 148:105095. [PMID: 34111479 DOI: 10.1016/j.neuint.2021.105095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 01/08/2023]
Abstract
Mitochondria are semi-autonomous organelle staging a crucial role in cellular stress response, energy metabolism and cell survival. Maintaining mitochondrial quality control is very important for its homeostasis. Pathological conditions such as oxidative stress and neurodegeneration, disrupt this quality control, and involvement of genetic and epigenetic materials in this disruption have been reported. These regulatory factors trigger mitochondrial imbalance, as seen in many neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, and Huntington's disease. The dynamic regulatory pathways i.e. mitophagy, biogenesis, permeability pore transitioning, fusion-fission are affected as a consequence and have been reviewed in this article. Moreover, several epigenetic mechanisms such as DNA methylation and histone modulation participating in such neurological disorders have also been discussed. Apart from it, therapeutic approaches targeting mitochondrial quality control have been tremendously explored showing ameliorative effects for these diseases, and have been discussed here with a novel perspective.
Collapse
Affiliation(s)
- Tanuja Ambekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Jyoti Pawar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Ramdev Rathod
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Monica Patel
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Rahul Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Shashi Bala Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
41
|
A Walk in the Memory, from the First Functional Approach up to Its Regulatory Role of Mitochondrial Bioenergetic Flow in Health and Disease: Focus on the Adenine Nucleotide Translocator. Int J Mol Sci 2021; 22:ijms22084164. [PMID: 33920595 PMCID: PMC8073645 DOI: 10.3390/ijms22084164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/11/2021] [Accepted: 04/16/2021] [Indexed: 12/19/2022] Open
Abstract
The mitochondrial adenine nucleotide translocator (ANT) plays the fundamental role of gatekeeper of cellular energy flow, carrying out the reversible exchange of ADP for ATP across the inner mitochondrial membrane. ADP enters the mitochondria where, through the oxidative phosphorylation process, it is the substrate of Fo-F1 ATP synthase, producing ATP that is dispatched from the mitochondrion to the cytoplasm of the host cell, where it can be used as energy currency for the metabolic needs of the cell that require energy. Long ago, we performed a method that allowed us to monitor the activity of ANT by continuously detecting the ATP gradually produced inside the mitochondria and exported in the extramitochondrial phase in exchange with externally added ADP, under conditions quite close to a physiological state, i.e., when oxidative phosphorylation takes place. More than 30 years after the development of the method, here we aim to put the spotlight on it and to emphasize its versatile applicability in the most varied pathophysiological conditions, reviewing all the studies, in which we were able to observe what really happened in the cell thanks to the use of the "ATP detecting system" allowing the functional activity of the ANT-mediated ADP/ATP exchange to be measured.
Collapse
|
42
|
Jaiquel Baron S, King MS, Kunji ER, Schirris TJ. Characterization of drug-induced human mitochondrial ADP/ATP carrier inhibition. Theranostics 2021; 11:5077-5091. [PMID: 33859735 PMCID: PMC8039944 DOI: 10.7150/thno.54936] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/18/2021] [Indexed: 01/10/2023] Open
Abstract
An increasing number of commonly prescribed drugs are known to interfere with mitochondrial function, causing cellular toxicity, but the underlying mechanisms are largely unknown. Although often not considered, mitochondrial transport proteins form a significant class of potential mitochondrial off-targets. So far, most drug interactions have been reported for the mitochondrial ADP/ATP carrier (AAC), which exchanges cytosolic ADP for mitochondrial ATP. Here, we show inhibition of cellular respiratory capacity by only a subset of the 18 published AAC inhibitors, which questions whether all compound do indeed inhibit such a central metabolic process. This could be explained by the lack of a simple, direct model system to evaluate and compare drug-induced AAC inhibition. Methods: For its development, we have expressed and purified human AAC1 (hAAC1) and applied two approaches. In the first, thermostability shift assays were carried out to investigate the binding of these compounds to human AAC1. In the second, the effect of these compounds on transport was assessed in proteoliposomes with reconstituted human AAC1, enabling characterization of their inhibition kinetics. Results: Of the proposed inhibitors, chebulinic acid, CD-437 and suramin are the most potent with IC50-values in the low micromolar range, whereas another six are effective at a concentration of 100 μM. Remarkably, half of all previously published AAC inhibitors do not show significant inhibition in our assays, indicating that they are false positives. Finally, we show that inhibitor strength correlates with a negatively charged surface area of the inhibitor, matching the positively charged surface of the substrate binding site. Conclusion: Consequently, we have provided a straightforward model system to investigate AAC inhibition and have gained new insights into the chemical compound features important for inhibition. Better evaluation methods of drug-induced inhibition of mitochondrial transport proteins will contribute to the development of drugs with an enhanced safety profile.
Collapse
Affiliation(s)
- Stephany Jaiquel Baron
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Martin S. King
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Edmund R.S. Kunji
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | - Tom J.J. Schirris
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
43
|
Valdés A, Lucio-Cazaña FJ, Castro-Puyana M, García-Pastor C, Fiehn O, Marina ML. Comprehensive metabolomic study of the response of HK-2 cells to hyperglycemic hypoxic diabetic-like milieu. Sci Rep 2021; 11:5058. [PMID: 33658594 PMCID: PMC7930035 DOI: 10.1038/s41598-021-84590-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
Diabetic nephropathy (DN) is the leading cause of chronic kidney disease. Although hyperglycaemia has been determined as the most important risk factor, hypoxia also plays a relevant role in the development of this disease. In this work, a comprehensive metabolomic study of the response of HK-2 cells, a human cell line derived from normal proximal tubular epithelial cells, to hyperglycemic, hypoxic diabetic-like milieu has been performed. Cells simultaneously exposed to high glucose (25 mM) and hypoxia (1% O2) were compared to cells in control conditions (5.5 mM glucose/18.6% O2) at 48 h. The combination of advanced metabolomic platforms (GC-TOF MS, HILIC- and CSH-QExactive MS/MS), freely available metabolite annotation tools, novel databases and libraries, and stringent cut-off filters allowed the annotation of 733 metabolites intracellularly and 290 compounds in the extracellular medium. Advanced bioinformatics and statistical tools demonstrated that several pathways were significantly altered, including carbohydrate and pentose phosphate pathways, as well as arginine and proline metabolism. Other affected metabolites were found in purine and lipid metabolism, the protection against the osmotic stress and the prevention of the activation of the β-oxidation pathway. Overall, the effects of the combined exposure of HK-cells to high glucose and hypoxia are reasonably compatible with previous in vivo works.
Collapse
Affiliation(s)
- Alberto Valdés
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España.
- West Coast Metabolomics Center, UC Davis, Davis, CA, USA.
| | - Francisco J Lucio-Cazaña
- Departamento de Biología de Sistemas, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España
| | - María Castro-Puyana
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España
- Instituto de Investigación Química Andrés M del Rio, IQAR, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España
| | - Coral García-Pastor
- Departamento de Biología de Sistemas, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España
| | - Oliver Fiehn
- West Coast Metabolomics Center, UC Davis, Davis, CA, USA
| | - María Luisa Marina
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España.
- Instituto de Investigación Química Andrés M del Rio, IQAR, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km. 33.600, 28871, Alcalá de Henares, Madrid, España.
| |
Collapse
|
44
|
Rabdosianone I, a Bitter Diterpene from an Oriental Herb, Suppresses Thymidylate Synthase Expression by Directly Binding to ANT2 and PHB2. Cancers (Basel) 2021; 13:cancers13050982. [PMID: 33652782 PMCID: PMC7956614 DOI: 10.3390/cancers13050982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 11/24/2022] Open
Abstract
Simple Summary In the present study, we found the novel pleiotropic regulation of the oncogene product thymidylate synthase (TS) by a chemical biology approach to identify rabdosianone I-binding proteins. Rabdosianone I, which is extracted from a traditional Asian herb Isodon japonicus Hara for longevity, suppressed TS expression at mRNA and protein levels. We immobilized rabdosianone I onto nano-magnetic beads and identified two mitochondrial proteins, adenine nucleotide translocase 2 (ANT2) and prohibitin 2 (PHB2), as the direct targets of rabdosianone I in cancer cells. Mechanistically, the knockdown of ANT2 or PHB2 promoted proteasomal degradation of the TS protein. In addition, PHB2 reduced TS mRNA levels. Thus, we provide previously unknown mechanisms of TS regulation by ANT2 and PHB2 and propose the possibility of rabdosianone I as a promising lead compound for the discovery of a novel TS suppressor. Abstract Natural products have numerous bioactivities and are expected to be a resource for potent drugs. However, their direct targets in cells often remain unclear. We found that rabdosianone I, which is a bitter diterpene from an oriental herb for longevity, Isodon japonicus Hara, markedly inhibited the growth of human colorectal cancer cells by downregulating the expression of thymidylate synthase (TS). Next, using rabdosianone I-immobilized nano-magnetic beads, we identified two mitochondrial inner membrane proteins, adenine nucleotide translocase 2 (ANT2) and prohibitin 2 (PHB2), as direct targets of rabdosianone I. Consistent with the action of rabdosianone I, the depletion of ANT2 or PHB2 reduced TS expression in a different manner. The knockdown of ANT2 or PHB2 promoted proteasomal degradation of TS protein, whereas that of not ANT2 but PHB2 reduced TS mRNA levels. Thus, our study reveals the ANT2- and PHB2-mediated pleiotropic regulation of TS expression and demonstrates the possibility of rabdosianone I as a lead compound of TS suppressor.
Collapse
|
45
|
Tsukahara T, Sahara Y, Ribeiro N, Tsukahara R, Gotoh M, Sakamoto S, Handa H, Murakami-Murofushi K. Adenine nucleotide translocase 2, a putative target protein for 2-carba cyclic phosphatidic acid in microglial cells. Cell Signal 2021; 82:109951. [PMID: 33592249 DOI: 10.1016/j.cellsig.2021.109951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/12/2021] [Accepted: 01/30/2021] [Indexed: 11/28/2022]
Abstract
Lipid-protein interactions play essential roles in many biological phenomena. Lysophospholipid mediators, such as cyclic phosphatidic acid (cPA), have been recognized as secondary messengers, yet few cellular targets for cPA have been identified to date. Furthermore, the molecular mechanism that activates these downstream signaling events remains unknown. In this study, using metabolically stabilized cPA carba-derivative (2ccPA)-immobilized magnetic beads, we identified adenine nucleotide translocase 2 (ANT2) as a 2ccPA-interacting protein in microglial cells. 2ccPA was tested for its ability to inhibit apoptosis caused by phenylarsine oxide in microglial cells. This damage was significantly improved upon 2ccPA treatment, along with cell proliferation, apoptosis, reactive oxygen species production, and intracellular ATP levels. This is the first report to suggest the direct binding of 2ccPA to ANT2 in microglial cells and provides evidence for a new benefit of 2ccPA in protecting microglial cells from apoptotic death induced by the ANT2-mediated signaling pathway.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Yasuka Sahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Ryoko Tsukahara
- Ochadai Academic Production, Ochanomizu University, Tokyo, Japan
| | - Mari Gotoh
- Institute for Human Life Innovation, Ochanomizu University, Tokyo, Japan
| | - Satoshi Sakamoto
- School of Life Science and Technology, Tokyo Institute of Technology, Kanagawa, Japan
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Tokyo, Japan
| | | |
Collapse
|
46
|
Ferver A, Greene E, Wideman R, Dridi S. Evidence of Mitochondrial Dysfunction in Bacterial Chondronecrosis With Osteomyelitis-Affected Broilers. Front Vet Sci 2021; 8:640901. [PMID: 33634182 PMCID: PMC7902039 DOI: 10.3389/fvets.2021.640901] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/08/2021] [Indexed: 11/13/2022] Open
Abstract
A leading cause of lameness in modern broilers is bacterial chondronecrosis with osteomyelitis (BCO). While it is known that the components of BCO are bacterial infection, necrosis, and inflammation, the mechanism behind BCO etiology is not yet fully understood. In numerous species, including chicken, mitochondrial dysfunction has been shown to have a role in the pathogenicity of numerous diseases. The mitochondria is a known target for intracellular bacterial infections, similar to that of common causative agents in BCO, as well as a known regulator of cellular metabolism, stress response, and certain types of cell death. This study aimed to determine the expression profile of genes involved in mitochondrial biogenesis, dynamics, and function. RNA was isolated form the tibias from BCO-affected and healthy broilers and used to measure target gene expression via real-time qPCR. Mitochondrial biogenesis factors PGC-1α and PGC-1β were both significantly upregulated in BCO along with mitochondrial fission factors OMA1, MTFR1, MTFP1, and MFF1 as well as cellular respiration-related genes FOXO3, FOXO4, and av-UCP. Conversely, genes involved in mitochondrial function, ANT, COXIV, and COX5A showed decreased mRNA levels in BCO-affected tibia. This study is the first to provide evidence of potential mitochondrial dysfunction in BCO bone and warrants further mechanistic investigation into how this dysfunction contributes to BCO etiology.
Collapse
Affiliation(s)
- Alison Ferver
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Elizabeth Greene
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Robert Wideman
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Sami Dridi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
47
|
Inhibitory effects of cynaropicrin and related sesquiterpene lactones from leaves of artichoke (Cynara scolymus L.) on induction of iNOS in RAW264.7 cells and its high-affinity proteins. J Nat Med 2021; 75:381-392. [PMID: 33484417 DOI: 10.1007/s11418-020-01479-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/25/2020] [Indexed: 12/15/2022]
Abstract
The methanolic extract of the leaves of artichoke (Cynara scolymus L.) was found to inhibit nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Among the constituents of the extract, six sesquiterpene lactones (cynaropicrin, grosheimin, 11β,13-dihydrocynaropicrin, 3β-hydroxy-8α-[(S)-3-hydroxy-2-methylpropionyloxy]guaia-4(15),10(14),11(13)-trien-1α,5α,6βH-12,6-olide, 3β-hydroxy-8α-[2-methoxymethyl-2-propenoyloxy]guaia-4(15),10(14),11(13)-trien-1α,5α,6βH-12,6-olide, and deacylcynaropicrin) inhibited NO production and/or inducible nitric oxide synthase (iNOS) induction. The acyl group having an α,β-unsaturated carbonyl group at the 8-position and the α-methylene-γ-butyrolactone moiety were important for the strong inhibitory activity. Our results suggested that these sesquiterpene lactones inhibited the LPS-induced iNOS expression via the suppression of the JAK-STAT signaling pathway in addition to the κNF-κB signaling pathway. With regard to the target molecules of the sesquiterpene lactones, high-affinity proteins of cynaropicrin were purified from the cell extract. ATP/ADP translocase 2 and tubulin were identified and suggested to be involved in the cytotoxic effects of cynaropicrin, although the target molecules for the inhibition of iNOS expression were not clarified.
Collapse
|
48
|
Mortalin depletion induces MEK/ERK-dependent and ANT/CypD-mediated death in vemurafenib-resistant B-Raf V600E melanoma cells. Cancer Lett 2021; 502:25-33. [PMID: 33440231 DOI: 10.1016/j.canlet.2020.12.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 01/07/2023]
Abstract
Therapy resistance to a selective B-Raf inhibitor (BRAFi) poses a challenge in treating patients with BRAF-mutant melanomas. Here, we report that RNA interference of mortalin (HSPA9/GRP75), a mitochondrial molecular chaperone often upregulated and mislocalized in melanoma, can effectively induce death of vemurafenib-resistant progenies of human B-RafV600E melanoma cell lines, A375 and Colo-829. Mortalin depletion induced death of vemurafenib-resistant cells at similar efficacy as observed in vemurafenib-naïve parental cells. This lethality was correlated with perturbed mitochondrial permeability and was attenuated by knockdown of adenine nucleotide translocase (ANT) and cyclophilin D (CypD), the key regulators of mitochondrial permeability. Chemical inhibition of MEK1/2 and ERK1/2 also suppressed mortalin depletion-induced death and mitochondrial permeability in these cells. These data suggest that mortalin and MEK/ERK regulate an ANT/CypD-associated mitochondrial death mechanism(s) in B-RafV600E melanoma cells and that this regulation is conserved even after these cells develop BRAFi resistance. We also show that doxycycline-induced mortalin depletion can effectively suppress the xenografts of vemurafenib-resistant A375 progeny in athymic nude mice. These findings suggest that mortalin has potential as a candidate therapeutic target for BRAFi-resistant BRAF-mutant tumors.
Collapse
|
49
|
Guo H, Chen T, Liang Z, Fan L, Shen Y, Zhou D. iTRAQ and PRM-based comparative proteomic profiling in gills of white shrimp Litopenaeus vannamei under copper stress. CHEMOSPHERE 2021; 263:128270. [PMID: 33297214 DOI: 10.1016/j.chemosphere.2020.128270] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 05/11/2023]
Abstract
Crustaceans are particularly sensitive to heavy metal pollution. Copper (Cu) is one of typical heavy metal pollutants in aquatic ecosystems. However, limited attention has been paid on the proteomic responses of shrimp under Cu stress. White shrimp Litopenaeus vannamei held in 5‰ seawater were exposed to 5 mg L-1 Cu for 3 h, and the regulatory mechanism in the gills was elucidated using iTRAQ-based quantitative proteomics. The results showed that a total of 5034 proteins were identified, 385 differentially expressed proteins (DEPs), including 147 differentially up-regulated proteins (DUPs) and 238 differentially down-regulated proteins (DDPs) were found. Bioinformatics analysis indicated the DEPs responding to Cu stress mainly involved in cytoskeleton, immune response, stress response, protein synthesis, detoxification, ion homeostasis and apoptosis. Furthermore, we still performed PRM analysis on sarcoplasmic calcium binding protein (SCP), serine proteinase inhibitor B3 (SPIB3), C-type lectin 4 (CTL4), cathepsin L (CATHL), JHE-like carboxylesterase 1 (CXE1) and paramyosin (PMY), and biochemical analysis on Cu/Zn-superoxide dismutase (Cu/Zn-SOD) to validate the iTRAQ results, respectively. The present proteome analysis revealed that Cu stress disrupted the ion homeostasis and protein synthesis, and L.vannamei mainly regulates a series of molecular pathways which contained many key proteins involved in the immune process to protect the organism from Cu stress. Our data provides more insight about the underlying mechanisms that related to the stress response of Cu exposure in crustacean.
Collapse
Affiliation(s)
- Hui Guo
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institute, Zhanjiang, China
| | - Tianci Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institute, Zhanjiang, China
| | - Zhi Liang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institute, Zhanjiang, China
| | - Lanfen Fan
- College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yuchun Shen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institute, Zhanjiang, China.
| | - Dayan Zhou
- Aquatic Species Introduction and Breeding Center of Guangxi Zhuang Autonomous Region, Nanning, 530031, China.
| |
Collapse
|
50
|
Trisolini L, Laera L, Favia M, Muscella A, Castegna A, Pesce V, Guerra L, De Grassi A, Volpicella M, Pierri CL. Differential Expression of ADP/ATP Carriers as a Biomarker of Metabolic Remodeling and Survival in Kidney Cancers. Biomolecules 2020; 11:38. [PMID: 33396658 PMCID: PMC7824283 DOI: 10.3390/biom11010038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/19/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
ADP/ATP carriers (AACs) are mitochondrial transport proteins playing a strategic role in maintaining the respiratory chain activity, fueling the cell with ATP, and also regulating mitochondrial apoptosis. To understand if AACs might represent a new molecular target for cancer treatment, we evaluated AAC expression levels in cancer/normal tissue pairs available on the Tissue Cancer Genome Atlas database (TCGA), observing that AACs are dysregulated in most of the available samples. It was observed that at least two AACs showed a significant differential expression in all the available kidney cancer/normal tissue pairs. Thus, we investigated AAC expression in the corresponding kidney non-cancer (HK2)/cancer (RCC-Shaw and CaKi-1) cell lines, grown in complete medium or serum starvation, for investigating how metabolic alteration induced by different growth conditions might influence AAC expression and resistance to mitochondrial apoptosis initiators, such as "staurosporine" or the AAC highly selective inhibitor "carboxyatractyloside". Our analyses showed that AAC2 and AAC3 transcripts are more expressed than AAC1 in all the investigated kidney cell lines grown in complete medium, whereas serum starvation causes an increase of at least two AAC transcripts in kidney cancer cell lines compared to non-cancer cells. However, the total AAC protein content is decreased in the investigated cancer cell lines, above all in the serum-free medium. The observed decrease in AAC protein content might be responsible for the decrease of OXPHOS activity and for the observed lowered sensitivity to mitochondrial apoptosis induced by staurosporine or carboxyatractyloside. Notably, the cumulative probability of the survival of kidney cancer patients seriously decreases with the decrease of AAC1 expression in KIRC and KIRP tissues making AAC1 a possible new biomarker of metabolic remodeling and survival in kidney cancers.
Collapse
Affiliation(s)
- Lucia Trisolini
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Luna Laera
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Maria Favia
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Antonella Muscella
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali (Di.S.Te.B.A.), Università del Salento, 73100 Lecce, Italy;
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Vito Pesce
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
- BROWSer S.r.l. c/o, Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70126 Bari, Italy
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
| | - Ciro Leonardo Pierri
- Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70125 Bari, Italy; (L.T.); (L.L.); (M.F.); (A.C.); (V.P.); (L.G.)
- BROWSer S.r.l. c/o, Department of Biosciences, Biotechnologies, Biopharmaceutics, University “Aldo Moro” of Bari, Via E. Orabona, 4, 70126 Bari, Italy
| |
Collapse
|