151
|
Xu B, Hu R, Liang Z, Chen T, Chen J, Hu Y, Jiang Y, Li Y. Metabolic regulation of the bone marrow microenvironment in leukemia. Blood Rev 2020; 48:100786. [PMID: 33353770 DOI: 10.1016/j.blre.2020.100786] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/24/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Most leukemia patients experience little benefit from immunotherapy, in part due to the immunosuppressive bone marrow microenvironment. Various metabolic mechanisms orchestrate the behaviors of immune cells and leukemia cells in the bone marrow microenvironment. Furthermore, leukemia cells regulate the bone marrow microenvironment through metabolism to generate an adequate supply of energy and to escape antitumor immune surveillance. Thus, the targeting of the interaction between leukemia cells and the bone marrow microenvironment provides a new therapeutic avenue. In this review, we describe the concept of the bone marrow microenvironment and several important metabolic processes of leukemia cells within the bone marrow microenvironment, including carbohydrate, lipid, and amino acid metabolism. In addition, we discuss how these metabolic pathways regulate antitumor immunity and reveal potential therapeutic targets.
Collapse
Affiliation(s)
- Binyan Xu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China
| | - Rong Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China
| | - Zhao Liang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China
| | - Tong Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Jianyu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China
| | - Yirong Jiang
- Department of Hematology, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, Guangdong 523059, PR China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, PR China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), 510005 Guangzhou, PR China.
| |
Collapse
|
152
|
Huang S, Petereit J, Millar AH. Loss of conserved mitochondrial CLPP and its functions lead to different phenotypes in plants and other organisms. PLANT SIGNALING & BEHAVIOR 2020; 15:1831789. [PMID: 33073672 PMCID: PMC7671067 DOI: 10.1080/15592324.2020.1831789] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Caseinolytic protease (CLPP) is an energy-dependent serine-type protease that plays a role in protein quality control. The CLPP gene is highly conserved across kingdoms and the protein is present in both bacteria and eukaryote organelles like mitochondria across a wide phylogenetic range. This pedigree has all the hallmarks of CLPP being an essential gene. However, in plants, disruption of mitochondrial CLPP has no impact on its growth, reminiscent of its nonessential role in some model fungi. Deletion of mitochondrial CLPP improves health and increased life span in the filamentous fungus, Podospora anserina, while loss of human mitochondrial CLPP leads to infertility and hearing loss. Recently it was revealed that both plant and human CLPP share a similar role in maintenance of the N-module of respiratory complex I. In addition, plant mitochondrial CLPP also coordinates the homeostasis of other mitochondrial protein complexes encoded by genes across mitochondrial and nuclear genomes. Understanding the contextual role of mitochondrial CLPP across kingdoms may help to understand these diverse sets of clpp phenotypes and the widespread conservation of CLPP genes.
Collapse
Affiliation(s)
- Shaobai Huang
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - Jakob Petereit
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
| | - A. Harvey Millar
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Australia
- CONTACT A. Harvey Millar ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley 6009, Perth, Western Australia
| |
Collapse
|
153
|
Prabhu VV, Morrow S, Rahman Kawakibi A, Zhou L, Ralff M, Ray J, Jhaveri A, Ferrarini I, Lee Y, Parker C, Zhang Y, Borsuk R, Chang WI, Honeyman JN, Tavora F, Carneiro B, Raufi A, Huntington K, Carlsen L, Louie A, Safran H, Seyhan AA, Tarapore RS, Schalop L, Stogniew M, Allen JE, Oster W, El-Deiry WS. ONC201 and imipridones: Anti-cancer compounds with clinical efficacy. Neoplasia 2020; 22:725-744. [PMID: 33142238 PMCID: PMC7588802 DOI: 10.1016/j.neo.2020.09.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/20/2022]
Abstract
ONC201 was originally discovered as TNF-Related Apoptosis Inducing Ligand (TRAIL)-inducing compound TIC10. ONC201 appears to act as a selective antagonist of the G protein coupled receptor (GPCR) dopamine receptor D2 (DRD2), and as an allosteric agonist of mitochondrial protease caseinolytic protease P (ClpP). Downstream of target engagement, ONC201 activates the ATF4/CHOP-mediated integrated stress response leading to TRAIL/Death Receptor 5 (DR5) activation, inhibits oxidative phosphorylation via c-myc, and inactivates Akt/ERK signaling in tumor cells. This typically results in DR5/TRAIL-mediated apoptosis of tumor cells; however, DR5/TRAIL-independent apoptosis, cell cycle arrest, or antiproliferative effects also occur. The effects of ONC201 extend beyond bulk tumor cells to include cancer stem cells, cancer associated fibroblasts and immune cells within the tumor microenvironment that can contribute to its efficacy. ONC201 is orally administered, crosses the intact blood brain barrier, and is under evaluation in clinical trials in patients with advanced solid tumors and hematological malignancies. ONC201 has single agent clinical activity in tumor types that are enriched for DRD2 and/or ClpP expression including specific subtypes of high-grade glioma, endometrial cancer, prostate cancer, mantle cell lymphoma, and adrenal tumors. Synergy with radiation, chemotherapy, targeted therapy and immune-checkpoint agents has been identified in preclinical models and is being evaluated in clinical trials. Structure-activity relationships based on the core pharmacophore of ONC201, termed the imipridone scaffold, revealed novel potent compounds that are being developed. Imipridones represent a novel approach to therapeutically target previously undruggable GPCRs, ClpP, and innate immune pathways in oncology.
Collapse
Key Words
- 5-fu, 5-fluorouracil
- a2a, adenosine 2a receptor
- alcl, anaplastic large cell lymphoma
- all, acute lymphoblastic leukemia
- aml, acute myeloid leukemia
- ampk, amp kinase
- atrt, atypical teratoid rhabdoid tumor
- auc, area under the curve
- brd, bromodomain
- camp, cyclic amp
- cck18, caspase-cleaved cytokeratin 18
- ck18, cytokeratin 18
- cll, chronic lymphocytic leukemia
- clpp, caseinolytic protease p
- clpx, caseinolytic mitochondrial matrix peptidase chaperone subunit x
- cml, chronic myelogenous leukemia
- crc, colorectal cancer
- csc, cancer stem cell
- ctcl, cutaneous t-cell lymphoma
- dipg, diffuse intrinsic pontine glioma
- dlbcl, diffuse large b-cell lymphoma
- dna-pkcs, dna-activated protein kinase catalytic subunit
- dr5, death receptor 5
- drd1, dopamine receptor d1
- drd2, dopamine receptor d2
- drd3, dopamine receptor d3
- drd4, dopamine receptor d4
- drd5, dopamine receptor d5
- dsrct, desmoplastic small round cell tumor
- ec, endometrial cancer
- egfr, epidermal growth factor receptor
- flair, fluid-attenuated inversion recovery
- gbm, glioblastoma multiforme
- gdsc, genomics of drug sensitivity in cancer
- girk, g protein-coupled inwardly rectifying potassium channel
- gnrh, gonadotropin-releasing hormone receptor
- gpcr, g protein coupled receptor
- hcc, hepatocellular carcinoma
- ihc, immunohistochemistry
- hgg, high-grade glioma
- isr, integrated stress response
- mcl, mantle cell lymphoma
- mm, multiple myeloma
- mtd, maximum tolerated dose
- nhl, non-hodgkin’s lymphoma
- nk, natural killer
- noael, no-observed-adverse-event-level
- nsclc, non-small cell lung cancer
- os, overall survival
- oxphos, oxidative phosphorylation
- pc-pg, pheochromocytoma-paraganglioma
- pd, pharmacodynamic
- pdx, patient-derived xenograft
- pfs, progression-free survival
- pk, pharmacokinetic
- plc, phospholipase c
- rano, response assessment in neuro-oncology
- recist, response evaluation criteria in solid tumors
- rhtrail, recombinant human trail
- rp2d, recommended phase ii dose
- sar, structure–activity relationship
- sclc, small-cell lung cancer
- tic10, trail-inducing compound 10
- tmz, temozolomide
- tnbc, triple-negative breast cancer
- trail, tnf-associated apoptosis-inducing ligand
- tunel, terminal deoxynucleotidyl transferase dutp nick end labeling
- who, world health organization
Collapse
Affiliation(s)
- Varun Vijay Prabhu
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA
| | - Sara Morrow
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA
| | | | - Lanlan Zhou
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Marie Ralff
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Jocelyn Ray
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Aakash Jhaveri
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Isacco Ferrarini
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Young Lee
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Cassandra Parker
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Yiqun Zhang
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Robyn Borsuk
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Wen-I Chang
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Joshua N Honeyman
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Fabio Tavora
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Benedito Carneiro
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Alexander Raufi
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Kelsey Huntington
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Lindsey Carlsen
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Anna Louie
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Howard Safran
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | - Attila A Seyhan
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA
| | | | - Lee Schalop
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA
| | - Martin Stogniew
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA
| | - Joshua E Allen
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA.
| | - Wolfgang Oster
- Oncoceutics, Inc., 3675 Market St, Suite 200, Philadelphia, PA 19104, USA
| | - Wafik S El-Deiry
- Warren Alpert Medical School, Brown University, 70 Ship Street, Room 537, Providence, RI 02912, USA.
| |
Collapse
|
154
|
Singh RP, Schimmer AD. Mitochondria regulate AML differentiation independent of oxidative phosphorylation and metabolism. Mol Cell Oncol 2020; 7:1815503. [PMID: 33241109 DOI: 10.1080/23723556.2020.1815503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Rashim Pal Singh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
155
|
Proximity labeling in mammalian cells with TurboID and split-TurboID. Nat Protoc 2020; 15:3971-3999. [PMID: 33139955 DOI: 10.1038/s41596-020-0399-0] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/18/2020] [Indexed: 12/19/2022]
Abstract
This protocol describes the use of TurboID and split-TurboID in proximity labeling applications for mapping protein-protein interactions and subcellular proteomes in live mammalian cells. TurboID is an engineered biotin ligase that uses ATP to convert biotin into biotin-AMP, a reactive intermediate that covalently labels proximal proteins. Optimized using directed evolution, TurboID has substantially higher activity than previously described biotin ligase-related proximity labeling methods, such as BioID, enabling higher temporal resolution and broader application in vivo. Split-TurboID consists of two inactive fragments of TurboID that can be reconstituted through protein-protein interactions or organelle-organelle interactions, which can facilitate greater targeting specificity than full-length enzymes alone. Proteins biotinylated by TurboID or split-TurboID are then enriched with streptavidin beads and identified by mass spectrometry. Here, we describe fusion construct design and characterization (variable timing), proteomic sample preparation (5-7 d), mass spectrometric data acquisition (2 d), and proteomic data analysis (1 week).
Collapse
|
156
|
Kim MS, Gernapudi R, Cedeño YC, Polster BM, Martinez R, Shapiro P, Kesari S, Nurmemmedov E, Passaniti A. Targeting breast cancer metabolism with a novel inhibitor of mitochondrial ATP synthesis. Oncotarget 2020; 11:3863-3885. [PMID: 33196708 PMCID: PMC7597410 DOI: 10.18632/oncotarget.27743] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 01/17/2023] Open
Abstract
Inhibitors of mitochondrial respiration and ATP synthesis may promote the selective killing of respiration-competent cancer cells that are critical for tumor progression. We previously reported that CADD522, a small molecule inhibitor of the RUNX2 transcription factor, has potential for breast cancer treatment. In the current study, we show that CADD522 inhibits mitochondrial oxidative phosphorylation by decreasing the mitochondrial oxygen consumption rate (OCR) and ATP production in human breast cancer cells in a RUNX2-independent manner. The enzyme activity of mitochondrial ATP synthase was inhibited by CADD522 treatment. Importantly, results from cellular thermal shift assays that detect drug-induced protein stabilization revealed that CADD522 interacts with both α and β subunits of the F1-ATP synthase complex. Differential scanning fluorimetry also demonstrated interaction of α subunits of the F1-ATP synthase to CADD522. These results suggest that CADD522 might target the enzymatic F1 subunits in the ATP synthase complex. CADD522 increased the levels of intracellular reactive oxygen species (ROS), which was prevented by MitoQ, a mitochondria-targeted antioxidant, suggesting that cancer cells exposed to CADD522 may elevate ROS from mitochondria. CADD522-increased mitochondrial ROS levels were enhanced by exogenously added pro-oxidants such as hydrogen peroxide or tert-butyl hydroperoxide. Conversely, CADD522-mediated cell growth inhibition was blocked by N-acetyl-l-cysteine, a general ROS scavenger. Therefore, CADD522 may exert its antitumor activity by increasing mitochondrial driven cellular ROS levels. Collectively, our data suggest in vitro proof-of-concept that supports inhibition of mitochondrial ATP synthase and ROS generation as contributors to the effectiveness of CADD522 in suppression of tumor growth.
Collapse
Affiliation(s)
- Myoung Sook Kim
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Ramkishore Gernapudi
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | | | - Brian M. Polster
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| | - Ramon Martinez
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Santosh Kesari
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Elmar Nurmemmedov
- John Wayne Cancer Institute and Pacific Neuroscience Institute at Providence Saint John’s Health Center, Santa Monica, CA, USA
| | - Antonino Passaniti
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, Baltimore, MD, USA
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
- Research Health Scientist, The Veteran's Health Administration Research & Development Service (VAMHCS), Baltimore, MD, USA
| |
Collapse
|
157
|
Stevens BM, Jones CL, Pollyea DA, Culp-Hill R, D'Alessandro A, Winters A, Krug A, Abbott D, Goosman M, Pei S, Ye H, Gillen AE, Becker MW, Savona MR, Smith C, Jordan CT. Fatty acid metabolism underlies venetoclax resistance in acute myeloid leukemia stem cells. ACTA ACUST UNITED AC 2020; 1:1176-1187. [PMID: 33884374 DOI: 10.1038/s43018-020-00126-z] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Venetoclax with azacitidine (ven/aza) has emerged as a promising regimen for acute myeloid leukemia (AML), with a high percentage of clinical remissions in newly diagnosed patients. However, approximately 30% of newly diagnosed and the majority of relapsed patients do not achieve remission with ven/aza. We previously reported that ven/aza efficacy is based on eradication of AML stem cells through a mechanism involving inhibition of amino acid metabolism, a process which is required in primitive AML cells to drive oxidative phosphorylation. Herein we demonstrate that resistance to ven/aza occurs via up-regulation of fatty acid oxidation (FAO), which occurs due to RAS pathway mutations, or as a compensatory adaptation in relapsed disease. Utilization of FAO obviates the need for amino acid metabolism, thereby rendering ven/aza ineffective. Pharmacological inhibition of FAO restores sensitivity to ven/aza in drug resistant AML cells. We propose inhibition of FAO as a therapeutic strategy to address ven/aza resistance.
Collapse
Affiliation(s)
- Brett M Stevens
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Courtney L Jones
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045
| | - Angelo D'Alessandro
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045.,Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045
| | - Amanda Winters
- Divsion of Pediatric Hematology and Oncology, University of Colorado Denver, Aurora, CO 80045
| | - Anna Krug
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Diana Abbott
- Department of Biostatistics and Informatics, University of Colorado Denver, Aurora, CO 80045
| | - Madeline Goosman
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Shanshan Pei
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Haobin Ye
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Austin E Gillen
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael W Becker
- Department of Medicine, James P. Wilmot Cancer Center, Rochester, NY, USA
| | - Michael R Savona
- Department of Internal Medicine, Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Clayton Smith
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| | - Craig T Jordan
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045
| |
Collapse
|
158
|
Nouri K, Feng Y, Schimmer AD. Mitochondrial ClpP serine protease-biological function and emerging target for cancer therapy. Cell Death Dis 2020; 11:841. [PMID: 33037181 PMCID: PMC7547079 DOI: 10.1038/s41419-020-03062-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Mitochondrial ClpP is a serine protease located in the mitochondrial matrix. This protease participates in mitochondrial protein quality control by degrading misfolded or damaged proteins, thus maintaining normal metabolic function. Mitochondrial ClpP is a stable heptamer ring with peptidase activity that forms a multimeric complex with the ATP-dependent unfoldase ClpX (ClpXP) leading to proteolytic activity. Emerging evidence demonstrates that ClpXP is over-expressed in hematologic malignancies and solid tumors and is necessary for the viability of a subset of tumors. In addition, both inhibition and hyperactivation of ClpXP leads to impaired respiratory chain activity and causes cell death in cancer cells. Therefore, targeting mitochondrial ClpXP could be a novel therapeutic strategy for the treatment of malignancy. Here, we review the structure and function of mitochondrial ClpXP as well as strategies to target this enzyme complex as a novel therapeutic approach for malignancy.
Collapse
Affiliation(s)
- Kazem Nouri
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yue Feng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
159
|
Carter JL, Hege K, Kalpage HA, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting mitochondrial respiration for the treatment of acute myeloid leukemia. Biochem Pharmacol 2020; 182:114253. [PMID: 33011159 DOI: 10.1016/j.bcp.2020.114253] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with variable presentation, molecular phenotype, and cytogenetic abnormalities and has seen very little improvement in patient survival over the last few decades. This heterogeneity supports poor prognosis partially through the variability in response to the standard chemotherapy. Further understanding of molecular heterogeneity has promoted the development of novel treatments, some of which target mitochondrial metabolism and function. This review discusses the relative dependency that AML cells have on mitochondrial function, and the ability to pivot this reliance to target important subsets of AML cells, including leukemia stem cells (LSCs). LSCs are tumor-initiating cells that are resistant to standard chemotherapy and promote the persistence and relapse of AML. Historically, LSCs have been targeted based on immunophenotype, but recent developments in the understanding of LSC metabolism has demonstrated unique abilities to target LSCs while sparing normal hematopoietic stem cells (HSCs) through inhibition of mitochondrial function. Here we highlight the use of small molecules that have been demonstrated to effectively target mitochondrial function. IACS-010759 and ME-344 target the electron transport chain (ETC) to inhibit oxidative phosphorylation (OXPHOS). The imipridone family (ONC201, ONC206, ONC212) of inhibitors target mitochondria through activation of ClpP mitochondrial protease and reduce function of essential pathways. These molecules offer a new mechanism for developing clinical therapies in AML and support novel strategies to target LSCs in parallel with conventional therapies.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Division of Pediatric Hematology and Oncology, Children's Hospital of Michigan, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
160
|
Zhang B, Li M, Yang W, Loor JJ, Liang Y, Wang S, Zhao Y, Guo H, Ma X, Yu L, Xu C. Mitochondrial dysfunction and endoplasmic reticulum stress in calf hepatocytes are associated with fatty acid-induced ORAI calcium release-activated calcium modulator 1 signaling. J Dairy Sci 2020; 103:11945-11956. [PMID: 32981726 DOI: 10.3168/jds.2020-18684] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022]
Abstract
The store-operated Ca2+ entry (SOCE) moiety ORAI calcium release-activated calcium modulator 1 (ORAI1) located in the endoplasmic reticulum (ER) participates in key cellular functions such as protein folding, transport, and secretion, and lipid metabolism. We used an in vitro approach to test whether exogenous fatty acids alter ORAI1 signaling and to explore potential consequences on mitochondrial dysfunction and ER stress. First, hepatocytes isolated from 4 healthy female calves (1 d old, 40-50 kg) were challenged with a 1.2 mM mixture of oleic, linoleic, palmitic, stearic, and palmitoleic acids for 0.5, 1, 3, 6, 9, and 12 h to measure oxidative stress [intracellular reduced glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), and hydrogen peroxide] and ER stress (protein abundance of PERK, IRE, ATF6, and GRP78). Concentrations of GSH and SOD decreased at 0.5 h, and MDA and hydrogen peroxide increased at 1 h; ER stress proteins increased at 6 h. To determine whether ER stress was caused by oxidative stress, primary calf hepatocytes were treated with the same 1.2 mM fatty acid mix or the reactive oxygen species (ROS) inhibitor N-acetylcysteine (NAC) for 6 h. We found that NAC prevented an increase in ER stress protein abundance. Next, the role of ORAI1 on ER stress was measured by transfecting hepatocytes with small interfering (si)ORAI1 or the ORAI1 inhibitor BTP2, followed by a challenge with 1.2 mM fatty acids for 3 h. Without inhibiting ORAI1, exogenous fatty acids upregulated ORAI1 mRNA and protein abundance, oxidative stress, ER stress proteins, and protein abundance of marker indicators of an opened mitochondrial permeability transition pore (mPTP). Inhibition with BPT2 or silencing via siORAI1 abrogated oxidative stress, including increased GSH concentration and SOD activity, decreased MDA, hydrogen peroxide, and ROS concentration; ER stress protein abundance was downregulated, and mitochondrial function was restored. Last, changes in markers of mPTP opening were evaluated by culturing hepatocytes for 6 h with the sarcoendoplasmic Ca2+ ATPase inhibitor thapsigargin or the calcium ionophore ionomycin. We detected an increase in VDAC1, CLPP, and CypD protein abundance, all of which indicated opening of the mPTP. Overall, data from these in vitro studies suggest that ORAI1 mediates ER stress induced by high concentrations of fatty acids, in part through alleviating mitochondrial dysfunction caused by oxidative stress.
Collapse
Affiliation(s)
- Bingbing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Ming Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Wei Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Yusheng Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Shuang Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Yingying Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Han Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Xinru Ma
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Liyun Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China
| | - Chuang Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, China.
| |
Collapse
|
161
|
Baudouin HCM, Pfeiffer L, Ochsenreiter T. A comparison of three approaches for the discovery of novel tripartite attachment complex proteins in Trypanosoma brucei. PLoS Negl Trop Dis 2020; 14:e0008568. [PMID: 32936798 PMCID: PMC7521757 DOI: 10.1371/journal.pntd.0008568] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 09/28/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022] Open
Abstract
Trypanosoma brucei is a single celled eukaryotic parasite and the causative agent of human African trypanosomiasis and nagana in cattle. Aside from its medical relevance, T. brucei has also been key to the discovery of several general biological principles including GPI-anchoring, RNA-editing and trans-splicing. The parasite contains a single mitochondrion with a singular genome. Recent studies have identified several molecular components of the mitochondrial genome segregation machinery (tripartite attachment complex, TAC), which connects the basal body of the flagellum to the mitochondrial DNA of T. brucei. The TAC component in closest proximity to the mitochondrial DNA is TAC102. Here we apply and compare three different approaches (proximity labelling, immunoprecipitation and yeast two-hybrid) to identify novel interactors of TAC102 and subsequently verify their localisation. Furthermore, we establish the direct interaction of TAC102 and p166 in the unilateral filaments of the TAC. Trypanosoma brucei belongs to a group of organisms that exist as human, animal and plant parasites. T. brucei (a human and animal parasite) has been developed as a model system to study basic biological as well as disease related questions in this group of organisms. We study how the parasite duplicates and divides its mitochondrial genome, an essential component of its energy generating machinery. The structure involved in dividing the mitochondrial genome into the daughter cells during cell division is called the tripartite attachment complex (TAC). The TAC is likely a unique structure not present in the host and thus might provide a new avenue for drug development. In this manuscript, we compare different techniques that allow the identification of novel components of this structure and verify the localisation of some of them. Furthermore, we also establish the interaction of two previously identified protein components.
Collapse
Affiliation(s)
- Hélène Clémentine Margareta Baudouin
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Laura Pfeiffer
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
162
|
Furukawa T, Katayama H, Oikawa A, Negishi L, Ichikawa T, Suzuki M, Murase K, Takayama S, Sakuda S. Dioctatin Activates ClpP to Degrade Mitochondrial Components and Inhibits Aflatoxin Production. Cell Chem Biol 2020; 27:1396-1409.e10. [PMID: 32888498 DOI: 10.1016/j.chembiol.2020.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/28/2020] [Accepted: 08/07/2020] [Indexed: 12/25/2022]
Abstract
Aflatoxin contamination of crops is a serious problem worldwide. Utilization of aflatoxin production inhibitors is attractive, as the elucidation of their modes of action contributes to clarifying the mechanism of aflatoxin production. Here, we identified mitochondrial protease ClpP as the target of dioctatin, an inhibitor of aflatoxin production of Aspergillus flavus. Dioctatin conferred uncontrolled caseinolytic capacity on ClpP of A. flavus and Escherichia coli. Dioctatin-bound ClpP selectively degraded mitochondrial energy-related proteins in vitro, including a subunit of respiratory chain complex V, which was also reduced by dioctatin in a ClpP-dependent manner in vivo. Dioctatin enhanced glycolysis and alcohol fermentation while reducing tricarboxylic acid cycle metabolites. These disturbances were accompanied by reduced histone acetylation and reduced expression of aflatoxin biosynthetic genes. Our results suggest that dioctatin inhibits aflatoxin production by inducing ClpP-mediated degradation of mitochondrial energy-related components, and that mitochondrial energy metabolism functions as a key determinant of aflatoxin production.
Collapse
Affiliation(s)
- Tomohiro Furukawa
- Department of Biosciences, Faculty of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya-shi, Tochigi 320-0003, Japan
| | - Hidekazu Katayama
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka-shi, Kanagawa 259-1292, Japan
| | - Akira Oikawa
- Department of Food, Life, and Environmental Sciences, Faculty of Agriculture, Yamagata University, 1-4-12 Kojirakawa-machi, Yamagata-shi, Yamagata 990-8560, Japan; RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-chou, Tsurumi-ku, Yokohama-shi, Kanagawa 230-0045, Japan
| | - Lumi Negishi
- Institute for Quantitative Biosciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takuma Ichikawa
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka-shi, Kanagawa 259-1292, Japan
| | - Michio Suzuki
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kohji Murase
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Seiji Takayama
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shohei Sakuda
- Department of Biosciences, Faculty of Science and Engineering, Teikyo University, 1-1 Toyosatodai, Utsunomiya-shi, Tochigi 320-0003, Japan.
| |
Collapse
|
163
|
Petereit J, Duncan O, Murcha MW, Fenske R, Cincu E, Cahn J, Pružinská A, Ivanova A, Kollipara L, Wortelkamp S, Sickmann A, Lee J, Lister R, Millar AH, Huang S. Mitochondrial CLPP2 Assists Coordination and Homeostasis of Respiratory Complexes. PLANT PHYSIOLOGY 2020; 184:148-164. [PMID: 32571844 PMCID: PMC7479914 DOI: 10.1104/pp.20.00136] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/12/2020] [Indexed: 05/04/2023]
Abstract
Protein homeostasis in eukaryotic organelles and their progenitor prokaryotes is regulated by a series of proteases including the caseinolytic protease (CLPP). CLPP has essential roles in chloroplast biogenesis and maintenance, but the significance of the plant mitochondrial CLPP remains unknown and factors that aid coordination of nuclear- and mitochondrial-encoded subunits for complex assembly in mitochondria await discovery. We generated knockout lines of the single gene for the mitochondrial CLP protease subunit, CLPP2, in Arabidopsis (Arabidopsis thaliana). Mutants showed a higher abundance of transcripts from mitochondrial genes encoding oxidative phosphorylation protein complexes, whereas nuclear genes encoding other subunits of the same complexes showed no change in transcript abundance. By contrast, the protein abundance of specific nuclear-encoded subunits in oxidative phosphorylation complexes I and V increased in CLPP2 knockouts, without accumulation of mitochondrial-encoded counterparts in the same complex. Complexes with subunits mainly or entirely encoded in the nucleus were unaffected. Analysis of protein import and function of complex I revealed that while function was retained, protein homeostasis was disrupted, leading to accumulation of soluble subcomplexes of nuclear-encoded subunits. Therefore, CLPP2 contributes to the mitochondrial protein degradation network through supporting coordination and homeostasis of protein complexes encoded across mitochondrial and nuclear genomes.
Collapse
Affiliation(s)
- Jakob Petereit
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Owen Duncan
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Monika W Murcha
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Ricarda Fenske
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Emilia Cincu
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Jonathan Cahn
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Adriana Pružinská
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Aneta Ivanova
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
| | - Stefanie Wortelkamp
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, Scotland, United Kingdom
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, D-44801 Bochum, Germany
| | - Jiwon Lee
- Centre for advanced Microscopy, The Australian National University, Acton, Australian Capital Territory 2601, Australia
| | - Ryan Lister
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
- The Harry Perkins Institute of Medical Research, Perth, Washington 6009, Australia
| | - A Harvey Millar
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| | - Shaobai Huang
- ARC Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Washington 6009, Australia
| |
Collapse
|
164
|
Jones CL, Stevens BM, Pollyea DA, Culp-Hill R, Reisz JA, Nemkov T, Gehrke S, Gamboni F, Krug A, Winters A, Pei S, Gustafson A, Ye H, Inguva A, Amaya M, Minhajuddin M, Abbott D, Becker MW, DeGregori J, Smith CA, D'Alessandro A, Jordan CT. Nicotinamide Metabolism Mediates Resistance to Venetoclax in Relapsed Acute Myeloid Leukemia Stem Cells. Cell Stem Cell 2020; 27:748-764.e4. [PMID: 32822582 DOI: 10.1016/j.stem.2020.07.021] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
Abstract
We previously demonstrated that leukemia stem cells (LSCs) in de novo acute myeloid leukemia (AML) patients are selectively reliant on amino acid metabolism and that treatment with the combination of venetoclax and azacitidine (ven/aza) inhibits amino acid metabolism, leading to cell death. In contrast, ven/aza fails to eradicate LSCs in relapsed/refractory (R/R) patients, suggesting altered metabolic properties. Detailed metabolomic analysis revealed elevated nicotinamide metabolism in relapsed LSCs, which activates both amino acid metabolism and fatty acid oxidation to drive OXPHOS, thereby providing a means for LSCs to circumvent the cytotoxic effects of ven/aza therapy. Genetic and pharmacological inhibition of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in nicotinamide metabolism, demonstrated selective eradication of R/R LSCs while sparing normal hematopoietic stem/progenitor cells. Altogether, these findings demonstrate that elevated nicotinamide metabolism is both the mechanistic basis for ven/aza resistance and a metabolic vulnerability of R/R LSCs.
Collapse
Affiliation(s)
- Courtney L Jones
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Brett M Stevens
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Sarah Gehrke
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Anna Krug
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Amanda Winters
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Shanshan Pei
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Annika Gustafson
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Haobin Ye
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Anagha Inguva
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Maria Amaya
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | | | - Diana Abbott
- Department of Biostatistics and Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Michael W Becker
- Department of Medicine, Division of Hematology/Oncology, University of Rochester, Rochester, NY 14627, USA
| | - James DeGregori
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Clayton A Smith
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA; Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO 80045, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
165
|
Mirali S, Schimmer AD. The role of mitochondrial proteases in leukemic cells and leukemic stem cells. Stem Cells Transl Med 2020; 9:1481-1487. [PMID: 32761807 PMCID: PMC7695628 DOI: 10.1002/sctm.20-0142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/18/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022] Open
Abstract
The biological function of most mitochondrial proteases has not been well characterized. Moreover, most of the available information on the normal function of these proteases has been derived from studies in model organisms. Recently, the mitochondrial proteases caseinolytic protease P (CLPP) and neurolysin (NLN) have been identified as therapeutic targets in acute myeloid leukemia (AML). Both proteases are overexpressed in approximately 40% of AML patients. Mechanistically, CLPP and NLN maintain the integrity of the mitochondrial respiratory chain: CLPP cleaves defective respiratory chain proteins, while NLN promotes the formation of respiratory chain supercomplexes. In this review, we highlight the functional consequences of inhibiting and activating mitochondrial proteases and discuss their potential as therapeutic targets in AML.
Collapse
Affiliation(s)
- Sara Mirali
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
166
|
Li J, Agarwal E, Bertolini I, Seo JH, Caino MC, Ghosh JC, Kossenkov AV, Liu Q, Tang HY, Goldman AR, Languino LR, Speicher DW, Altieri DC. The mitophagy effector FUNDC1 controls mitochondrial reprogramming and cellular plasticity in cancer cells. Sci Signal 2020; 13:13/642/eaaz8240. [PMID: 32723812 DOI: 10.1126/scisignal.aaz8240] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria are signaling hubs in eukaryotic cells. Here, we showed that the mitochondrial FUN14 domain-containing protein-1 (FUNDC1), an effector of Parkin-independent mitophagy, also participates in cellular plasticity by sustaining oxidative bioenergetics, buffering ROS production, and supporting cell proliferation. Targeting this pathway in cancer cells suppressed tumor growth but rendered transformed cells more motile and invasive in a manner dependent on ROS-mediated mitochondrial dynamics and mitochondrial repositioning to the cortical cytoskeleton. Global metabolomics and proteomics profiling identified a FUNDC1 interactome at the mitochondrial inner membrane, comprising the AAA+ protease, LonP1, and subunits of oxidative phosphorylation, complex V (ATP synthase). Independently of its previously identified role in mitophagy, FUNDC1 enabled LonP1 proteostasis, which in turn preserved complex V function and decreased ROS generation. Therefore, mitochondrial reprogramming by a FUNDC1-LonP1 axis controls tumor cell plasticity by switching between proliferative and invasive states in cancer.
Collapse
Affiliation(s)
- Jie Li
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Ekta Agarwal
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Irene Bertolini
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Jae Ho Seo
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - M Cecilia Caino
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jagadish C Ghosh
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Qin Liu
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Hsin-Yao Tang
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Aaron R Goldman
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - David W Speicher
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA.,Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA 19104, USA.,Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, PA 19104, USA. .,Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA 19104, USA
| |
Collapse
|
167
|
Xu M, Seneviratne AK, Schimmer AD. Phospholipid metabolism regulates AML growth and stemness. Aging (Albany NY) 2020; 11:3895-3897. [PMID: 31237862 PMCID: PMC6628983 DOI: 10.18632/aging.102055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Mingjing Xu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ayesh K Seneviratne
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
168
|
St-Germain JR, Samavarchi Tehrani P, Wong C, Larsen B, Gingras AC, Raught B. Variability in Streptavidin-Sepharose Matrix Quality Can Significantly Affect Proximity-Dependent Biotinylation (BioID) Data. J Proteome Res 2020; 19:3554-3561. [PMID: 32628020 DOI: 10.1021/acs.jproteome.0c00117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Due to their ease of use and high binding affinity, streptavidin-based purification tools have become widely used for isolating biotinylated compounds from complex mixtures. We and others routinely use streptavidin-sepharose matrices to isolate biotinylated polypeptides generated in proximity-dependent biotinylation approaches, such as BioID or APEX. However, we noted sporadic, substantial variation in the quality of BioID experiments performed in the same laboratories over time, using seemingly identical protocols. Identifying the source of this problem, here, we highlight considerable variability in streptavidin contamination derived from different production lots of streptavidin-sepharose beads from the same manufacturer and demonstrate that high levels of streptavidin peptide contamination can have detrimental effects on BioID data. We also describe two simple, rapid approaches to assess the degree of streptavidin "shedding" from individual lots of the sepharose matrix before use to avoid the use of lower quality reagent.
Collapse
Affiliation(s)
- Jonathan R St-Germain
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Payman Samavarchi Tehrani
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Department of Molecular Genetics, University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Cassandra Wong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Department of Molecular Genetics, University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Brett Larsen
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Department of Molecular Genetics, University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, and Department of Molecular Genetics, University of Toronto, 600 University Avenue, Toronto, ON M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
169
|
Kim J, Cheong JH. Role of Mitochondria-Cytoskeleton Interactions in the Regulation of Mitochondrial Structure and Function in Cancer Stem Cells. Cells 2020; 9:cells9071691. [PMID: 32674438 PMCID: PMC7407978 DOI: 10.3390/cells9071691] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 12/16/2022] Open
Abstract
Despite the promise of cancer medicine, major challenges currently confronting the treatment of cancer patients include chemoresistance and recurrence. The existence of subpopulations of cancer cells, known as cancer stem cells (CSCs), contributes to the failure of cancer therapies and is associated with poor clinical outcomes. Of note, one of the recently characterized features of CSCs is augmented mitochondrial function. The cytoskeleton network is essential in regulating mitochondrial morphology and rearrangement, which are inextricably linked to its functions, such as oxidative phosphorylation (OXPHOS). The interaction between the cytoskeleton and mitochondria can enable CSCs to adapt to challenging conditions, such as a lack of energy sources, and to maintain their stemness. Cytoskeleton-mediated mitochondrial trafficking and relocating to the high energy requirement region are crucial steps in epithelial-to-mesenchymal transition (EMT). In addition, the cytoskeleton itself interplays with and blocks the voltage-dependent anion channel (VDAC) to directly regulate bioenergetics. In this review, we describe the regulation of cellular bioenergetics in CSCs, focusing on the cytoskeleton-mediated dynamic control of mitochondrial structure and function.
Collapse
Affiliation(s)
- Jungmin Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Jae-Ho Cheong
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Korea
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2228-2094; Fax: +82-2-313-8289
| |
Collapse
|
170
|
Botham A, Coyaud E, Nirmalanandhan VS, Gronda M, Hurren R, Maclean N, St-Germain J, Mirali S, Laurent E, Raught B, Schimmer A. Global Interactome Mapping of Mitochondrial Intermembrane Space Proteases Identifies a Novel Function for HTRA2. Proteomics 2020; 19:e1900139. [PMID: 31617661 DOI: 10.1002/pmic.201900139] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/30/2019] [Indexed: 12/20/2022]
Abstract
A number of unique proteases localize to specific sub-compartments of the mitochondria, but the functions of these enzymes are poorly defined. Here, in vivo proximity-dependent biotinylation (BioID) is used to map the interactomes of seven proteases localized to the mitochondrial intermembrane space (IMS). In total, 802 high confidence proximity interactions with 342 unique proteins are identified. While all seven proteases co-localized with the IMS markers OPA1 and CLPB, 230 of the interacting partners are unique to just one or two protease bait proteins, highlighting the ability of BioID to differentiate unique interactomes within the confined space of the IMS. Notably, high-temperature requirement peptidase 2 (HTRA2) interacts with eight of 13 components of the mitochondrial intermembrane space bridging (MIB) complex, a multiprotein assembly essential for the maintenance of mitochondrial cristae structure. Knockdown of HTRA2 disrupts cristae in HEK 293 and OCI-AML2 cells, and leads to increased intracellular levels of the MIB subunit IMMT. Using a cell-free assay it is demonstrated that HTRA2 can degrade recombinant IMMT but not two other core MIB complex subunits, SAMM50 and CHCHD3. The IMS protease interactome thus represents a rich dataset that can be mined to uncover novel IMS protease biology.
Collapse
Affiliation(s)
- Aaron Botham
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, ON, Canada
| | - Etienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | | | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | - Neil Maclean
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | - Jonathan St-Germain
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | - Sara Mirali
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, M5G 1L7, ON, Canada
| | - Estelle Laurent
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, ON, Canada
| | - Aaron Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, M5G 1L7, ON, Canada
| |
Collapse
|
171
|
Dittmar D, Reder A, Schlüter R, Riedel K, Hecker M, Gerth U. Complementation studies with human ClpP in Bacillus subtilis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118744. [PMID: 32442436 DOI: 10.1016/j.bbamcr.2020.118744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/29/2022]
Abstract
ATP-dependent intracellular proteolysis is essential for all living organisms. ClpP, the proteolytic subunit of the ATP-dependent Clp proteases, shares 56% protein identity between B. subtilis and man. The aim of this study was to verify, whether human ClpP (HClpP) is able to substitute the bacterial pendant, BClpP, irrespectively of the huge evolutionary distance. For this reason hclpP was expressed from the natural B. subtilis promoters at the original chromosomal site. Growth at 37 °C as well as sporulation in the presence of hclpP depict an intermediate phenotype between wild type and clpP mutant suggesting a partial functional substitution of BClpP by HClpP. Northern as well as Western blot analyses show a similar induction pattern of both, bclpP and hclpP during heat stress on the mRNA as well as on the protein levels. Co-immunoprecipitation experiments imply specific interaction of HClpP with bacterial ClpC, ClpX and ClpE during control as well as heat stress conditions. Radioactive pulse-chase labeling and immunoprecipitation revealed that a ClpXP substrate, the short-living regulatory protein MgsR, is degraded by HClpP, although with an extremely slower rate in comparison to BClpP. The occurrence of an exceptional thickened cell wall of a clpP mutant can be almost fully reversed by the complementation with HClpP. The utilization of the HClpP expressing strain as a test system for new biological or synthetic active substances targeting BClpP is discussed.
Collapse
Affiliation(s)
- Denise Dittmar
- Institute of Microbiology of the Department of Biology, University of Greifswald, Germany; Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Reder
- Institute of Microbiology of the Department of Biology, University of Greifswald, Germany; Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Rabea Schlüter
- Imaging Center of the Department of Biology, University of Greifswald, Germany
| | - Katharina Riedel
- Institute of Microbiology of the Department of Biology, University of Greifswald, Germany
| | - Michael Hecker
- Institute of Microbiology of the Department of Biology, University of Greifswald, Germany
| | - Ulf Gerth
- Institute of Microbiology of the Department of Biology, University of Greifswald, Germany.
| |
Collapse
|
172
|
Abstract
We recently identified the mitochondrial peptidase, neurolysin (NLN), as a top hit in an acute myeloid leukemia (AML) viability screen. Using chemical and genetic approaches, we demonstrated that loss of NLN disrupted respiratory chain supercomplex assembly and impaired oxidative metabolism in AML. Moreover, inhibition of NLN in vitro and in vivo reduced the growth of AML cells.
Collapse
Affiliation(s)
- Sara Mirali
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Medicine, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.,Department of Medicine, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
173
|
O'Malley J, Kumar R, Inigo J, Yadava N, Chandra D. Mitochondrial Stress Response and Cancer. Trends Cancer 2020; 6:688-701. [PMID: 32451306 DOI: 10.1016/j.trecan.2020.04.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/16/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022]
Abstract
Cancer cells survive and adapt to many types of stress including hypoxia, nutrient deprivation, metabolic, and oxidative stress. These stresses are sensed by diverse cellular signaling processes, leading to either degradation of mitochondria or alleviation of mitochondrial stress. This review discusses signaling during sensing and mitigation of stress involving mitochondrial communication with the endoplasmic reticulum, and how retrograde signaling upregulates the mitochondrial stress response to maintain mitochondrial integrity. The importance of the mitochondrial unfolded protein response, an emerging pathway that alleviates cellular stress, will be elaborated with respect to cancer. Detailed understanding of cellular pathways will establish mitochondrial stress response as a key mechanism for cancer cell survival leading to cancer progression and resistance, and provide a potential therapeutic target in cancer.
Collapse
Affiliation(s)
- Jordan O'Malley
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rahul Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Joseph Inigo
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nagendra Yadava
- Department of Anesthesiology and Center for Shock, Trauma, and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
174
|
Singh RP, Jeyaraju DV, Voisin V, Hurren R, Xu C, Hawley JR, Barghout SH, Khan DH, Gronda M, Wang X, Jitkova Y, Sharon D, Liyanagae S, MacLean N, Seneviratene AK, Mirali S, Borenstein A, Thomas GE, Soriano J, Orouji E, Minden MD, Arruda A, Chan SM, Bader GD, Lupien M, Schimmer AD. Disrupting Mitochondrial Copper Distribution Inhibits Leukemic Stem Cell Self-Renewal. Cell Stem Cell 2020; 26:926-937.e10. [PMID: 32416059 DOI: 10.1016/j.stem.2020.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 02/27/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022]
Abstract
Leukemic stem cells (LSCs) rely on oxidative metabolism and are differentially sensitive to targeting mitochondrial pathways, which spares normal hematopoietic cells. A subset of mitochondrial proteins is folded in the intermembrane space via the mitochondrial intermembrane assembly (MIA) pathway. We found increased mRNA expression of MIA pathway substrates in acute myeloid leukemia (AML) stem cells. Therefore, we evaluated the effects of inhibiting this pathway in AML. Genetic and chemical inhibition of ALR reduces AML growth and viability, disrupts LSC self-renewal, and induces their differentiation. ALR inhibition preferentially decreases its substrate COX17, a mitochondrial copper chaperone, and knockdown of COX17 phenocopies ALR loss. Inhibiting ALR and COX17 increases mitochondrial copper levels which in turn inhibit S-adenosylhomocysteine hydrolase (SAHH) and lower levels of S-adenosylmethionine (SAM), DNA methylation, and chromatin accessibility to lower LSC viability. These results provide insight into mechanisms through which mitochondrial copper controls epigenetic status and viability of LSCs.
Collapse
Affiliation(s)
- Rashim Pal Singh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Danny V Jeyaraju
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Rose Hurren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Changjiang Xu
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - James R Hawley
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Samir H Barghout
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Dilshad H Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Marcela Gronda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xiaoming Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yulia Jitkova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - David Sharon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sanduni Liyanagae
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Neil MacLean
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Sara Mirali
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Adina Borenstein
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Geethu E Thomas
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Joelle Soriano
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Elias Orouji
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Andrea Arruda
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gary D Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
175
|
Yang N, Cao Q, Hu S, Xu C, Fan K, Chen F, Yang C, Liang H, Wu M, Bae T, Lan L. Alteration of protein homeostasis mediates the interaction of
Pseudomonas aeruginosa
with
Staphylococcus aureus. Mol Microbiol 2020; 114:423-442. [DOI: 10.1111/mmi.14519] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Nana Yang
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Qiao Cao
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
- College of Life Science Northwest University Xi'an China
| | - Shuyang Hu
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Chenchen Xu
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Ke Fan
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Feifei Chen
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
- College of Life Science Northwest University Xi'an China
| | - Cai‐Guang Yang
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
| | - Haihua Liang
- College of Life Science Northwest University Xi'an China
| | - Min Wu
- Department of Biomedical Sciences University of North Dakota Grand Forks ND USA
| | - Taeok Bae
- Department of Microbiology and Immunology Indiana University School of Medicine‐Northwest Gary IN USA
| | - Lefu Lan
- University of Chinese Academy of Sciences Beijing China
- State Key Laboratory of Drug Research Shanghai Institute of Materia Medica Chinese Academy of Sciences Shanghai China
- College of Life Science Northwest University Xi'an China
- NMPA Key Laboratory for Testing Technology of Pharmaceutical Microbiology Shanghai Institute for Food and Drug Control Shanghai China
| |
Collapse
|
176
|
Wang SF, Chen S, Tseng LM, Lee HC. Role of the mitochondrial stress response in human cancer progression. Exp Biol Med (Maywood) 2020; 245:861-878. [PMID: 32326760 PMCID: PMC7268930 DOI: 10.1177/1535370220920558] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPACT STATEMENT Dysregulated mitochondria often occurred in cancers. Mitochondrial dysfunction might contribute to cancer progression. We reviewed several mitochondrial stresses in cancers. Mitochondrial stress responses might contribute to cancer progression. Several mitochondrion-derived molecules (ROS, Ca2+, oncometabolites, exported mtDNA, mitochondrial double-stranded RNA, humanin, and MOTS-c), integrated stress response, and mitochondrial unfolded protein response act as retrograde signaling pathways and might be critical in the development and progression of cancer. Targeting these mitochondrial stress responses may be an important strategy for cancer treatment.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, 112 Taipei
- School of Pharmacy, Taipei Medical University, 110 Taipei
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, 112 Taipei
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, CA 91010, USA
| | - Ling-Ming Tseng
- Division of General Surgery, Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, 112 Taipei
- Department of Surgery, School of Medicine, National Yang-Ming University, 112 Taipei
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, 112 Taipei
| |
Collapse
|
177
|
Kalvala AK, Khan I, Gundu C, Kumar A. An Overview on ATP Dependent and Independent Proteases Including an Anterograde to Retrograde Control on Mitochondrial Function; Focus on Diabetes and Diabetic Complications. Curr Pharm Des 2020; 25:2584-2594. [PMID: 31317835 DOI: 10.2174/1381612825666190718153901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/12/2019] [Indexed: 12/17/2022]
Abstract
Mitochondria are the central power stations of the cell involved with a myriad of cell signalling pathways that contribute for whole health status of the cell. It is a well known fact that not only mitochondrial genome encodes for mitochondrial proteins but there are several other mitochondrial specific proteins encoded by nuclear genome which regulate plethora of cell catabolic and anabolic process. Anterograde pathways include nuclear gene encoded proteins and their specific transport into the mitochondria and regulation of mitochondrial homeostasis. The retrograde pathways include crosstalk between the mitochondria and cytoplasmic proteins. Indeed, ATP dependent and independent proteases are identified to be very critical in balancing anterograde to retrograde signalling and vice versa to maintain the cell viability or cell death. Different experimental studies conducted on silencing the genes of these proteases have shown embryonic lethality, cancer cells death, increased hepatic glucose output, insulin tolerance, increased protein exclusion bodies, mitochondrial dysfunction, and defect in mitochondrial biogenesis, increased inflammation, Apoptosis etc. These experimental studies included from eubacteria to eukaryotes. Hence, many lines of theories proposed these proteases are conservative from eubacteria to eukaryotes. However, the regulation of these proteases at gene level is not clearly understood and still research is warranted. In this review, we articulated the origin and regulation of these proteases and the cross talk between the nucleus and mitochondria vice versa, and highlighted the role of these proteases in diabetes and diabetic complications in human diseases.
Collapse
Affiliation(s)
- Anil Kumar Kalvala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Bala Nagar, India
| | - Islauddin Khan
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Bala Nagar, India
| | - Chayanika Gundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Bala Nagar, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Bala Nagar, India
| |
Collapse
|
178
|
Sha Z, Fishovitz J, Wang S, Chilakala S, Xu Y, Lee I. A Selective Fluorogenic Peptide Substrate for the Human Mitochondrial ATP-Dependent Protease Complex ClpXP. Chembiochem 2020; 21:2037-2048. [PMID: 32180333 DOI: 10.1002/cbic.202000030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/01/2020] [Indexed: 11/07/2022]
Abstract
The goal of this work is to identify differences in the substrate determinants of two human mitochondrial matrix ATP-dependent proteases, human ClpXP (hClpXP) and human Lon (hLon). This information allows the generation of protease-specific peptide substrates that can be used as chemical biology tools to investigate the physiological functions of hClpXP. These enzymes play a role in protein quality control, but currently the physiological functions of human ClpXP are not well defined. In this study, the degradation profile of casein, an alanine positional scanning decapeptide library, and a specific peptide sequence found in an endogenous substrate of bacterial ClpXP by hClpXP as well as hLon were examined. Based on our findings, we generated a specific fluorogenic peptide substrate, FR-Cleptide, for hClpXP with a kcat of 2.44±0.15 s-1 and Km =262±43 μM, respectively. The FR-Cleptide substrate was successfully used to identify a leucine methyl ketone as a potent lead inhibitor, and to detect endogenous hClpXP activity in HeLa cell lysate. We propose that the fluorogenic peptide substrate is a valuable tool for quantitatively monitoring the activity of hClpXP in cell lysate, as well as mechanistic characterization of hClpXP. The peptide-based chemical tools developed in this study will complement the substrates developed for human Lon in aiding the investigation of the physiological functions of the respective protease.
Collapse
Affiliation(s)
- Zhou Sha
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Jennifer Fishovitz
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana, 46556, USA
| | - Susan Wang
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Sujatha Chilakala
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, 44115, USA.,Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Beverly Hills, CA, 90211, USA
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, 44115, USA
| | - Irene Lee
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
179
|
Sha Z, Chilakala S, Crabill G, Cheng I, Xu Y, Fishovitz J, Lee I. A Proteolytic Site-Directed Affinity Label to Inhibit the Human ATP-Dependent Protease Caseinolytic Complex XP. Chembiochem 2020; 21:2049-2059. [PMID: 32180302 DOI: 10.1002/cbic.202000031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/01/2020] [Indexed: 11/10/2022]
Abstract
Human caseinolytic protease component X and P (hClpXP) is a heterooligomeric ATP-dependent protease. The hClpX subunit catalyzes ATP hydrolysis whereas the hClpP subunit catalyzes peptide bond cleavage. In this study, we generated a peptidyl chloromethyl ketone (dansyl-FAPAL-CMK) that inhibited the hClpP subunit through alkylation of the catalytic His122, which was detected by LC-MS. This inhibitor is composed of a peptide sequence derived from a hydrolyzed peptide product of a substrate cleaved by hClpXP. Binding of FAPAL positions the electrophilic chloromethyl ketone moiety near His122 where alkylation occurs. Dansyl FAPAL-CMK exhibits selectivity for hClpXP over other ATP-dependent proteases such as hLon and the 26S proteasome and abolishes hClpXP activity in HeLa cell lysate. Using the fluorogenic peptide substrate FR-Cleptide as reporter, we detected biphasic inhibition time courses; this supports a slow-binding, time-dependent, covalent inhibition mechanism that is often found in active-site directed affinity labels. Because this inhibitor reacts only with hClpXP but not hLon or the proteasome, it has the potential to serve as a chemical tool to help validate endogenous protein substrates of hClpXP in cell lysate, thereby benefiting investigation of the physiological functions of hClpXP in different cell types or tissue samples.
Collapse
Affiliation(s)
- Zhou Sha
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Sujatha Chilakala
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, 44115, USA.,Present address: Lawrence J. Ellison Institute for Transformative Medicine of USC, University of Southern California, Beverly Hills, CA, 90211, USA
| | - George Crabill
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA.,Present address: University of Maryland School of Medicine, Baltimore, MD, 21202, USA
| | - Iteen Cheng
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA.,Present address: Agilent Technologies, Cleveland, OH, 44106, USA
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, 44115, USA
| | - Jennifer Fishovitz
- Department of Chemistry and Physics, Saint Mary's College, Notre Dame, Indiana, 46556, USA
| | - Irene Lee
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
180
|
Pei S, Pollyea DA, Gustafson A, Stevens BM, Minhajuddin M, Fu R, Riemondy KA, Gillen AE, Sheridan RM, Kim J, Costello JC, Amaya ML, Inguva A, Winters A, Ye H, Krug A, Jones CL, Adane B, Khan N, Ponder J, Schowinsky J, Abbott D, Hammes A, Myers JR, Ashton JM, Nemkov T, D'Alessandro A, Gutman JA, Ramsey HE, Savona MR, Smith CA, Jordan CT. Monocytic Subclones Confer Resistance to Venetoclax-Based Therapy in Patients with Acute Myeloid Leukemia. Cancer Discov 2020; 10:536-551. [PMID: 31974170 PMCID: PMC7124979 DOI: 10.1158/2159-8290.cd-19-0710] [Citation(s) in RCA: 323] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/03/2019] [Accepted: 01/17/2020] [Indexed: 12/12/2022]
Abstract
Venetoclax-based therapy can induce responses in approximately 70% of older previously untreated patients with acute myeloid leukemia (AML). However, up-front resistance as well as relapse following initial response demonstrates the need for a deeper understanding of resistance mechanisms. In the present study, we report that responses to venetoclax +azacitidine in patients with AML correlate closely with developmental stage, where phenotypically primitive AML is sensitive, but monocytic AML is more resistant. Mechanistically, resistant monocytic AML has a distinct transcriptomic profile, loses expression of venetoclax target BCL2, and relies on MCL1 to mediate oxidative phosphorylation and survival. This differential sensitivity drives a selective process in patients which favors the outgrowth of monocytic subpopulations at relapse. Based on these findings, we conclude that resistance to venetoclax + azacitidine can arise due to biological properties intrinsic to monocytic differentiation. We propose that optimal AML therapies should be designed so as to independently target AML subclones that may arise at differing stages of pathogenesis. SIGNIFICANCE: Identifying characteristics of patients who respond poorly to venetoclax-based therapy and devising alternative therapeutic strategies for such patients are important topics in AML. We show that venetoclax resistance can arise due to intrinsic molecular/metabolic properties of monocytic AML cells and that such properties can potentially be targeted with alternative strategies.
Collapse
Affiliation(s)
- Shanshan Pei
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel A Pollyea
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Annika Gustafson
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Mohammad Minhajuddin
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Rui Fu
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado
| | - Kent A Riemondy
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado
| | - Austin E Gillen
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado
| | - Ryan M Sheridan
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado
| | - Jihye Kim
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - James C Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Maria L Amaya
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado
| | - Anagha Inguva
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Amanda Winters
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Haobin Ye
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Anna Krug
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Courtney L Jones
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Biniam Adane
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Nabilah Khan
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jessica Ponder
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jeffrey Schowinsky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado
| | - Diana Abbott
- Center for Innovative Design and Analysis, Colorado School of Public Health, Aurora, Colorado
| | - Andrew Hammes
- Center for Innovative Design and Analysis, Colorado School of Public Health, Aurora, Colorado
| | - Jason R Myers
- Genomics Research Center, University of Rochester, Rochester, New York
| | - John M Ashton
- Genomics Research Center, University of Rochester, Rochester, New York
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Angelo D'Alessandro
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Jonathan A Gutman
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Haley E Ramsey
- Department of Internal Medicine, Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Michael R Savona
- Department of Internal Medicine, Vanderbilt University School of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Clayton A Smith
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
181
|
Luo J, Zeng B, Tao C, Lu M, Ren G. ClpP regulates breast cancer cell proliferation, invasion and apoptosis by modulating the Src/PI3K/Akt signaling pathway. PeerJ 2020; 8:e8754. [PMID: 32195060 PMCID: PMC7069407 DOI: 10.7717/peerj.8754] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Caseinolytic protease P (ClpP), which is located on the inner mitochondrial membrane, degrades mitochondrial proteins damaged by oxidative stress. The role of ClpP varies among tumor types. However, the expression pattern and biological functions of ClpP in breast cancer (BC) have not yet been investigated. METHODS The Cancer Genome Atlas (TCGA) and Kaplan Meier-plotter database were used to analyze the expression level of ClpP in BC tissues, relationships with clinicopathological characteristics, and the influence on the prognosis of BC. Protein and mRNA expression levels of ClpP in BC cell lines and tissues were detected by quantitative real-time PCR, western blot and immunohistochemical (IHC) analyses. The colony formation assay, transwell assay and flow cytometric analysis were performed to assess various functions of ClpP. Western blot analysis was also conducted to determine the mechanism of ClpP. RESULTS ClpP expression was markedly increased in BC cells and tissues. High expression of ClpP was significantly correlated with the T stage, estrogen receptor (ER) expression, and poor recurrence-free survival (RFS) in TCGA and Kaplan Meier-plotter database. ClpP silencing significantly inhibited proliferation, migration, invasion, and promoted apoptosis of BC cells, which resulted in suppression of the Src/PI3K/Akt signaling pathway. The gain-of-function assay confirmed partial these results.
Collapse
Affiliation(s)
- Juan Luo
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Beilei Zeng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunfang Tao
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengqi Lu
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
182
|
Imipridone Anticancer Compounds Ectopically Activate the ClpP Protease and Represent a New Scaffold for Antibiotic Development. Genetics 2020; 214:1103-1120. [PMID: 32094149 PMCID: PMC7153937 DOI: 10.1534/genetics.119.302851] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/06/2020] [Indexed: 11/18/2022] Open
Abstract
The imipridones ONC201 and ONC212 selectively kill cancer cells and have been ascribed multiple mechanisms-of-action. Genome-wide CRISPR knockout screens revealed that loss of the mitochondrial proteases CLPP and MIPEP confer strong resistance to both compounds... Systematic genetic interaction profiles can reveal the mechanisms-of-action of bioactive compounds. The imipridone ONC201, which is currently in cancer clinical trials, has been ascribed a variety of different targets. To investigate the genetic dependencies of imipridone action, we screened a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) knockout library in the presence of either ONC201 or its more potent analog ONC212. Loss of the mitochondrial matrix protease CLPP or the mitochondrial intermediate peptidase MIPEP conferred strong resistance to both compounds. Biochemical and surrogate genetic assays showed that impridones directly activate CLPP and that MIPEP is necessary for proteolytic maturation of CLPP into a catalytically competent form. Quantitative proteomic analysis of cells treated with ONC212 revealed degradation of many mitochondrial as well as nonmitochondrial proteins. Prompted by the conservation of ClpP from bacteria to humans, we found that the imipridones also activate ClpP from Escherichia coli, Bacillus subtilis, and Staphylococcus aureus in biochemical and genetic assays. ONC212 and acyldepsipeptide-4 (ADEP4), a known activator of bacterial ClpP, caused similar proteome-wide degradation profiles in S. aureus. ONC212 suppressed the proliferation of a number of Gram-positive (S. aureus, B. subtilis, and Enterococcus faecium) and Gram-negative species (E. coli and Neisseria gonorrhoeae). Moreover, ONC212 enhanced the ability of rifampin to eradicate antibiotic-tolerant S. aureus persister cells. These results reveal the genetic dependencies of imipridone action in human cells and identify the imipridone scaffold as a new entry point for antibiotic development.
Collapse
|
183
|
Deshwal S, Fiedler KU, Langer T. Mitochondrial Proteases: Multifaceted Regulators of Mitochondrial Plasticity. Annu Rev Biochem 2020; 89:501-528. [PMID: 32075415 DOI: 10.1146/annurev-biochem-062917-012739] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria are essential metabolic hubs that dynamically adapt to physiological demands. More than 40 proteases residing in different compartments of mitochondria, termed mitoproteases, preserve mitochondrial proteostasis and are emerging as central regulators of mitochondrial plasticity. These multifaceted enzymes limit the accumulation of short-lived, regulatory proteins within mitochondria, modulate the activity of mitochondrial proteins by protein processing, and mediate the degradation of damaged proteins. Various signaling cascades coordinate the activity of mitoproteases to preserve mitochondrial homeostasis and ensure cell survival. Loss of mitoproteases severely impairs the functional integrity of mitochondria, is associated with aging, and causes pleiotropic diseases. Understanding the dual function of mitoproteases as regulatory and quality control enzymes will help unravel the role of mitochondrial plasticity in aging and disease.
Collapse
Affiliation(s)
- Soni Deshwal
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany;
| | - Kai Uwe Fiedler
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany;
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany; .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
184
|
Gupta A, Ökesli-Armlovich A, Morgens D, Bassik MC, Khosla C. A genome-wide analysis of targets of macrolide antibiotics in mammalian cells. J Biol Chem 2020; 295:2057-2067. [PMID: 31915244 DOI: 10.1074/jbc.ra119.010770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 01/05/2020] [Indexed: 01/04/2023] Open
Abstract
Macrolide antibiotics, such as erythromycin and josamycin, are natural polyketide products harboring 14- to 16-membered macrocyclic lactone rings to which various sugars are attached. These antibiotics are used extensively in the clinic because of their ability to inhibit bacterial protein synthesis. More recently, some macrolides have been shown to also possess anti-inflammatory and other therapeutic activities in mammalian cells. To better understand the targets and effects of this drug class in mammalian cells, we used a genome-wide shRNA screen in K562 cancer cells to identify genes that modulate cellular sensitivity to josamycin. Among the most sensitizing hits were proteins involved in mitochondrial translation and the mitochondrial unfolded protein response, glycolysis, and the mitogen-activated protein kinase signaling cascade. Further analysis revealed that cells treated with josamycin or other antibacterial agents exhibited impaired oxidative phosphorylation and metabolic shifts to glycolysis. Interestingly, we observed that knockdown of the mitogen-activated protein kinase kinase kinase 4 (MAP3K4) gene, which contributes to p38 mitogen-activated protein kinase signaling, sensitized cells only to josamycin but not to other antibacterial agents. There is a growing interest in better characterizing the therapeutic effects and toxicities of antibiotics in mammalian cells to guide new applications in both cellular and clinical studies. To our knowledge, this is the first report of an unbiased genome-wide screen to investigate the effects of a clinically used antibiotic on human cells.
Collapse
Affiliation(s)
- Amita Gupta
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305
| | - Aye Ökesli-Armlovich
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Chemistry, Stanford University, Stanford, California 94305
| | - David Morgens
- Department of Genetics, Stanford University, Stanford, California 94305
| | - Michael C Bassik
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Genetics, Stanford University, Stanford, California 94305
| | - Chaitan Khosla
- Department of Chemical Engineering, Stanford University, Stanford, California 94305; Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, California 94305; Department of Chemistry, Stanford University, Stanford, California 94305; Department of Biochemistry, Stanford University, Stanford, California 94305.
| |
Collapse
|
185
|
Lin HH, Yu M, Sriramoju MK, Hsu STD, Liu CT, Lai EM. A High-Throughput Interbacterial Competition Screen Identifies ClpAP in Enhancing Recipient Susceptibility to Type VI Secretion System-Mediated Attack by Agrobacterium tumefaciens. Front Microbiol 2020; 10:3077. [PMID: 32117077 PMCID: PMC7012810 DOI: 10.3389/fmicb.2019.03077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022] Open
Abstract
The type VI secretion system (T6SS) is an effector delivery system used by Gram-negative bacteria to kill other bacteria or eukaryotic hosts to gain fitness. The plant pathogen Agrobacterium tumefaciens utilizes its T6SS to kill other bacteria, such as Escherichia coli. We observed that the A. tumefaciens T6SS-dependent killing outcome differs when using different T6SS-lacking, K-12 E. coli strains as a recipient cell. Thus, we hypothesized that the A. tumefaciens T6SS killing outcome not only relies on the T6SS activity of the attacker cells but also depends on the recipient cells. Here, we developed a high-throughput interbacterial competition platform to test the hypothesis by screening for mutants with reduced killing outcomes caused by A. tumefaciens strain C58. Among the 3,909 strains in the E. coli Keio library screened, 16 mutants with less susceptibility to A. tumefaciens C58 T6SS-dependent killing were identified, and four of them were validated by complementation test. Among the four, the clpP encoding ClpP protease, which is universal and highly conserved in both prokaryotes and eukaryotic organelles, was selected for further characterizations. We demonstrated that ClpP is responsible for enhancing susceptibility to the T6SS killing. Because ClpP protease depends on other adapter proteins such as ClpA and ClpX for substrate recognition, further mutant studies followed by complementation tests were carried out to reveal that ClpP-associated AAA+ ATPase ClpA, but not ClpX, is involved in enhancing susceptibility to A. tumefaciens T6SS killing. Moreover, functional and biochemical studies of various ClpP amino acid substitution variants provided evidence that ClpA–ClpP interaction is critical in enhancing susceptibility to the T6SS killing. This study highlights the importance of recipient factors in determining the outcome of the T6SS killing and shows the universal ClpP protease as a novel recipient factor hijacked by the T6SS of A. tumefaciens.
Collapse
Affiliation(s)
- Hsiao-Han Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan.,Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Manda Yu
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | | | | | - Chi-Te Liu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Erh-Min Lai
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
186
|
Rochford G, Molphy Z, Kavanagh K, McCann M, Devereux M, Kellett A, Howe O. Cu(ii) phenanthroline–phenazine complexes dysregulate mitochondrial function and stimulate apoptosis. Metallomics 2020; 12:65-78. [DOI: 10.1039/c9mt00187e] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Herein we report the central role of the mitochondria in the cytotoxicity of four developmental cytotoxic copper(ii) complexes [Cu(phen)2]2+, [Cu(DPQ)(Phen)]2+, [Cu(DPPZ)(Phen)]2+and [Cu(DPPN)(Phen)]2+superior to cisplatin and independent of resistance in a range of cells.
Collapse
Affiliation(s)
- Garret Rochford
- FOCAS Research Institute and School of Biological & Health Sciences
- Technological University Dublin
- Dublin 8
- Ireland
| | - Zara Molphy
- School of Chemical Science and The National Institute for Cellular Biotechnology
- Dublin City University
- Dublin 9
- Ireland
| | | | - Malachy McCann
- Department of Chemistry
- Maynooth University
- Maynooth
- Ireland
| | - Michael Devereux
- FOCAS Research Institute and School of Biological & Health Sciences
- Technological University Dublin
- Dublin 8
- Ireland
| | - Andrew Kellett
- School of Chemical Science and The National Institute for Cellular Biotechnology
- Dublin City University
- Dublin 9
- Ireland
| | - Orla Howe
- FOCAS Research Institute and School of Biological & Health Sciences
- Technological University Dublin
- Dublin 8
- Ireland
| |
Collapse
|
187
|
Batsivari A, Haltalli MLR, Passaro D, Pospori C, Lo Celso C, Bonnet D. Dynamic responses of the haematopoietic stem cell niche to diverse stresses. Nat Cell Biol 2020; 22:7-17. [PMID: 31907409 DOI: 10.1038/s41556-019-0444-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/27/2019] [Indexed: 01/01/2023]
Abstract
Adult haematopoietic stem cells (HSCs) mainly reside in the bone marrow, where stromal and haematopoietic cells regulate their function. The steady state HSC niche has been extensively studied. In this Review, we focus on how bone marrow microenvironment components respond to different insults including inflammation, malignant haematopoiesis and chemotherapy. We highlight common and unique patterns among multiple cell types and their environment and discuss current limitations in our understanding of this complex and dynamic tissue.
Collapse
Affiliation(s)
- Antoniana Batsivari
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
| | - Myriam Luydmila Rachelle Haltalli
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK
- Lo Celso Laboratory, The Francis Crick Institute, London, UK
| | - Diana Passaro
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
| | - Constandina Pospori
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK
- Lo Celso Laboratory, The Francis Crick Institute, London, UK
| | - Cristina Lo Celso
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK.
- Department of Life Sciences, Imperial College London, South Kensington campus, London, UK.
- Lo Celso Laboratory, The Francis Crick Institute, London, UK.
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute , London, UK.
| |
Collapse
|
188
|
Abstract
In this Review, Rashkovan et al. discuss the role of cancer metabolic circuitries feeding anabolism and redox potential in leukemia development and recent progress in translating these important findings to the clinic. Leukemia cell proliferation requires up-regulation and rewiring of metabolic pathways to feed anabolic cell growth. Oncogenic drivers directly and indirectly regulate metabolic pathways, and aberrant metabolism is central not only for leukemia proliferation and survival, but also mediates oncogene addiction with significant implications for the development of targeted therapies. This review explores leukemia metabolic circuitries feeding anabolism, redox potential, and energy required for tumor propagation with an emphasis on emerging therapeutic opportunities.
Collapse
Affiliation(s)
- Marissa Rashkovan
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA.,Department of Pediatrics, Columbia University, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
189
|
Venkatesh S, Suzuki CK. Cell stress management by the mitochondrial LonP1 protease - Insights into mitigating developmental, oncogenic and cardiac stress. Mitochondrion 2019; 51:46-61. [PMID: 31756517 DOI: 10.1016/j.mito.2019.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/24/2019] [Accepted: 10/02/2019] [Indexed: 11/15/2022]
Abstract
Mitochondrial LonP1 is an essential stress response protease that mediates mitochondrial proteostasis, metabolism and bioenergetics. Homozygous and compound heterozygous variants in the LONP1 gene encoding the LonP1 protease have recently been shown to cause a diverse spectrum of human pathologies, ranging from classical mitochondrial disease phenotypes, profound neurologic impairment and multi-organ dysfunctions, some of which are uncommon to mitochondrial disorders. In this review, we focus primarily on human LonP1 and discuss findings, which demonstrate its multidimensional roles in maintaining mitochondrial proteostasis and adapting cells to metabolic flux and stress during normal physiology and disease processes. We also discuss emerging roles of LonP1 in responding to developmental, oncogenic and cardiac stress.
Collapse
Affiliation(s)
- Sundararajan Venkatesh
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Carolyn K Suzuki
- Department of Microbiology, Biochemistry & Molecular Genetics, New Jersey Medical School - Rutgers, The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|
190
|
Wong KS, Houry WA. Chemical Modulation of Human Mitochondrial ClpP: Potential Application in Cancer Therapeutics. ACS Chem Biol 2019; 14:2349-2360. [PMID: 31241890 DOI: 10.1021/acschembio.9b00347] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The human ClpP proteolytic complex (HsClpP) is a serine protease located in the mitochondrial matrix and participates in the maintenance of the mitochondrial proteome among other cellular functions. HsClpP typically forms a multimeric complex with the AAA+ protein unfoldase HsClpX. Notably, compared to that of normal, healthy cells, the expression of HsClpP in many types of solid and nonsolid cancers is found to be upregulated. While the exact role of HsClpP in tumorigenesis is not clear, certain types of cancers are highly dependent on the protease for cell proliferation and metastasis. In light of these observations, recent research has focused on the discovery and characterization of small organic molecules that can target and modulate HsClpP activity. These include compounds that inhibit HsClpP's proteolytic activity via covalent modification of its catalytic Ser residue as well as those that activate and dysregulate HsClpP by displacing HsClpX to negate its regulatory role. Importantly, several of these compounds have been shown to induce HsClpP-dependent apoptotic cell death in a variety of cancerous cells. This review provides an overview of these research efforts and highlights the various types of small molecule modulators of HsClpP activity with respect to their potential use as cancer therapeutics.
Collapse
Affiliation(s)
- Keith S. Wong
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Walid A. Houry
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1M1, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada
| |
Collapse
|
191
|
Santin YG. Uncovering the In Vivo Proxisome Using Proximity‐Tagging Methods. Bioessays 2019; 41:e1900131. [DOI: 10.1002/bies.201900131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/04/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Yoann G. Santin
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires, Institut de Microbiologie de la MéditerranéeAix‐Marseille Université – CNRS UMR7255 31 Chemin Joseph Aiguier, CS70071, 13402 Marseille Cedex 09 France
| |
Collapse
|
192
|
Gatsogiannis C, Balogh D, Merino F, Sieber SA, Raunser S. Cryo-EM structure of the ClpXP protein degradation machinery. Nat Struct Mol Biol 2019; 26:946-954. [PMID: 31582852 PMCID: PMC6783313 DOI: 10.1038/s41594-019-0304-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/19/2019] [Indexed: 12/23/2022]
Abstract
The ClpXP machinery is a two component protease complex performing
targeted protein degradation in bacteria and mitochondria. The complex consists
of the AAA+ chaperone ClpX and the peptidase ClpP. The hexameric ClpX utilizes
the energy of ATP binding and hydrolysis to engage, unfold and translocate
substrates into the catalytic chamber of tetradecameric ClpP where they are
degraded. Formation of the complex involves a symmetry mismatch, since hexameric
AAA+ rings bind axially to the opposing stacked heptameric rings of the
tetradecameric ClpP. Here we present the cryo-EM structure of ClpXP from
Listeria monocytogenes. We unravel the heptamer-hexamer
binding interface and provide novel insights into the ClpX-ClpP crosstalk and
activation mechanism. The comparison with available crystal structures of ClpP
and ClpX in different states allows us to understand important aspects of
ClpXP’s complex mode of action and provides a structural framework for
future pharmacological applications.
Collapse
Affiliation(s)
- Christos Gatsogiannis
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Dora Balogh
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Stephan A Sieber
- Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
193
|
Tiosano D, Mears JA, Buchner DA. Mitochondrial Dysfunction in Primary Ovarian Insufficiency. Endocrinology 2019; 160:2353-2366. [PMID: 31393557 PMCID: PMC6760336 DOI: 10.1210/en.2019-00441] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
Primary ovarian insufficiency (POI) is defined by the loss or dysfunction of ovarian follicles associated with amenorrhea before the age of 40. Symptoms include hot flashes, sleep disturbances, and depression, as well as reduced fertility and increased long-term risk of cardiovascular disease. POI occurs in ∼1% to 2% of women, although the etiology of most cases remains unexplained. Approximately 10% to 20% of POI cases are due to mutations in a single gene or a chromosomal abnormality, which has provided considerable molecular insight into the biological underpinnings of POI. Many of the genes for which mutations have been associated with POI, either isolated or syndromic cases, function within mitochondria, including MRPS22, POLG, TWNK, LARS2, HARS2, AARS2, CLPP, and LRPPRC. Collectively, these genes play roles in mitochondrial DNA replication, gene expression, and protein synthesis and degradation. Although mutations in these genes clearly implicate mitochondrial dysfunction in rare cases of POI, data are scant as to whether these genes in particular, and mitochondrial dysfunction in general, contribute to most POI cases that lack a known etiology. Further studies are needed to better elucidate the contribution of mitochondria to POI and determine whether there is a common molecular defect in mitochondrial function that distinguishes mitochondria-related genes that when mutated cause POI vs those that do not. Nonetheless, the clear implication of mitochondrial dysfunction in POI suggests that manipulation of mitochondrial function represents an important therapeutic target for the treatment or prevention of POI.
Collapse
Affiliation(s)
- Dov Tiosano
- Division of Pediatric Endocrinology, Ruth Rappaport Children’s Hospital, Rambam Medical Center, Haifa, Israel
- Rappaport Family Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Jason A Mears
- Center for Mitochondrial Diseases, Case Western Reserve University, Cleveland, Ohio
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio
- Research Institute for Children’s Health, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
194
|
Mattes K, Vellenga E, Schepers H. Differential redox-regulation and mitochondrial dynamics in normal and leukemic hematopoietic stem cells: A potential window for leukemia therapy. Crit Rev Oncol Hematol 2019; 144:102814. [PMID: 31593878 DOI: 10.1016/j.critrevonc.2019.102814] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/12/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
The prognosis for many patients with acute myeloid leukemia (AML) is poor, mainly due to disease relapse driven by leukemia stem cells (LSCs). Recent studies have highlighted the unique metabolic properties of LSCs, which might represent opportunities for LSC-selective targeting. LSCs characteristically have low levels of reactive oxygen species (ROS), which apparently result from a combination of low mitochondrial activity and high activity of ROS-removing pathways such as autophagy. Due to this low activity, LSCs are highly dependent on mitochondrial regulatory mechanisms. These include the anti-apoptotic protein BCL-2, which also has crucial roles in regulating the mitochondrial membrane potential, and proteins involved in mitophagy. Here we review the different pathways that impact mitochondrial activity and redox-regulation, and highlight their relevance for the functionality of both HSCs and LSCs. Additionally, novel AML therapy strategies that are based on interference with those pathways, including the promising BCL-2 inhibitor Venetoclax, are summarized.
Collapse
Affiliation(s)
- Katharina Mattes
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Edo Vellenga
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hein Schepers
- Department of Hematology, Cancer Research Center Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
195
|
Key J, Kohli A, Bárcena C, López-Otín C, Heidler J, Wittig I, Auburger G. Global Proteome of LonP1+/- Mouse Embryonal Fibroblasts Reveals Impact on Respiratory Chain, but No Interdependence between Eral1 and Mitoribosomes. Int J Mol Sci 2019; 20:E4523. [PMID: 31547314 PMCID: PMC6770551 DOI: 10.3390/ijms20184523] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Research on healthy aging shows that lifespan reductions are often caused by mitochondrial dysfunction. Thus, it is very interesting that the deletion of mitochondrial matrix peptidase LonP1 was observed to abolish embryogenesis, while deletion of the mitochondrial matrix peptidase Caseinolytic Mitochondrial Matrix Peptidase Proteolytic Subunit (ClpP) prolonged survival. To unveil the targets of each enzyme, we documented the global proteome of LonP1+/- mouse embryonal fibroblasts (MEF), for comparison with ClpP-/- depletion. Proteomic profiles of LonP1+/- MEF generated by label-free mass spectrometry were further processed with the STRING (Search tool for the retrieval of interacting genes) webserver Heidelberg for protein interactions. ClpP was previously reported to degrade Eral1 as a chaperone involved in mitoribosome assembly, so ClpP deficiency triggers the accumulation of mitoribosomal subunits and inefficient translation. LonP1+/- MEF also showed Eral1 accumulation, but no systematic effect on mitoribosomal subunits. In contrast to ClpP-/- profiles, several components of the respiratory complex-I membrane arm, of the glutathione pathway and of lysosomes were accumulated, whereas the upregulation of numerous innate immune defense components was similar. Overall, LonP1, as opposed to ClpP, appears to have no effect on translational machinery, instead it shows enhanced respiratory dysfunction; this agrees with reports on the human CODAS syndrome (syndrome with cerebral, ocular, dental, auricular, and skeletal anomalies) caused by LonP1 mutations.
Collapse
Affiliation(s)
- Jana Key
- Experimental Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany.
| | - Aneesha Kohli
- Experimental Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany.
| | - Clea Bárcena
- Departamento de Bioquimica y Biologia Molecular, Facultad de Medicina, Instituto Universitario de Oncologia (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain.
| | - Carlos López-Otín
- Departamento de Bioquimica y Biologia Molecular, Facultad de Medicina, Instituto Universitario de Oncologia (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain.
| | - Juliana Heidler
- Functional Proteomics Group, Goethe-University Hospital, 60590 Frankfurt am Main, Germany.
| | - Ilka Wittig
- Functional Proteomics Group, Goethe-University Hospital, 60590 Frankfurt am Main, Germany.
| | - Georg Auburger
- Experimental Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
196
|
Felix J, Weinhäupl K, Chipot C, Dehez F, Hessel A, Gauto DF, Morlot C, Abian O, Gutsche I, Velazquez-Campoy A, Schanda P, Fraga H. Mechanism of the allosteric activation of the ClpP protease machinery by substrates and active-site inhibitors. SCIENCE ADVANCES 2019; 5:eaaw3818. [PMID: 31517045 PMCID: PMC6726451 DOI: 10.1126/sciadv.aaw3818] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 08/02/2019] [Indexed: 05/14/2023]
Abstract
Coordinated conformational transitions in oligomeric enzymatic complexes modulate function in response to substrates and play a crucial role in enzyme inhibition and activation. Caseinolytic protease (ClpP) is a tetradecameric complex, which has emerged as a drug target against multiple pathogenic bacteria. Activation of different ClpPs by inhibitors has been independently reported from drug development efforts, but no rationale for inhibitor-induced activation has been hitherto proposed. Using an integrated approach that includes x-ray crystallography, solid- and solution-state nuclear magnetic resonance, molecular dynamics simulations, and isothermal titration calorimetry, we show that the proteasome inhibitor bortezomib binds to the ClpP active-site serine, mimicking a peptide substrate, and induces a concerted allosteric activation of the complex. The bortezomib-activated conformation also exhibits a higher affinity for its cognate unfoldase ClpX. We propose a universal allosteric mechanism, where substrate binding to a single subunit locks ClpP into an active conformation optimized for chaperone association and protein processive degradation.
Collapse
Affiliation(s)
- Jan Felix
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Katharina Weinhäupl
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Christophe Chipot
- LPCT, UMR 7019 Université de Lorraine CNRS, Vandoeuvre-les-Nancy F-54500, France
- Laboratoire International Associé CNRS and University of Illinois at Urbana−Champaign, Vandoeuvre-les-Nancy F-54506, France
- Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana, IL 61801, USA
| | - François Dehez
- LPCT, UMR 7019 Université de Lorraine CNRS, Vandoeuvre-les-Nancy F-54500, France
- Laboratoire International Associé CNRS and University of Illinois at Urbana−Champaign, Vandoeuvre-les-Nancy F-54506, France
| | - Audrey Hessel
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Diego F. Gauto
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Cecile Morlot
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Olga Abian
- Institute of Biocomputation and Physics of Complex Systems (BIFI), Joint Units IQFR-CSIC-BIFI and GBsC-CSIC-BIFI, and Department of Biochemistry and Molecular and Cell Biology, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), 50009 Zaragoza, Spain
- Biomedical Research Networking Centre for Liver and Digestive Diseases (CIBERehd), Madrid, Spain
- Aragon Health Sciences Institute (IACS), 50009 Zaragoza, Spain
| | - Irina Gutsche
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
| | - Adrian Velazquez-Campoy
- Institute of Biocomputation and Physics of Complex Systems (BIFI), Joint Units IQFR-CSIC-BIFI and GBsC-CSIC-BIFI, and Department of Biochemistry and Molecular and Cell Biology, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), 50009 Zaragoza, Spain
- Biomedical Research Networking Centre for Liver and Digestive Diseases (CIBERehd), Madrid, Spain
- Fundacion ARAID, Government of Aragon, 50018 Zaragoza, Spain
| | - Paul Schanda
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
- Corresponding author. (H.F.); (P.S.)
| | - Hugo Fraga
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS, IBS, 71 Avenue des Martyrs, F-38044 Grenoble, France
- Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Corresponding author. (H.F.); (P.S.)
| |
Collapse
|
197
|
Noncanonical mitochondrial unfolded protein response impairs placental oxidative phosphorylation in early-onset preeclampsia. Proc Natl Acad Sci U S A 2019; 116:18109-18118. [PMID: 31439814 PMCID: PMC6731647 DOI: 10.1073/pnas.1907548116] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Preeclampsia endangers the lives and well-being of mother and baby. The syndrome is associated with placental dysfunction. High demand for energy to support active nutrient transport and hormone production increases placental susceptibility to mitochondrial stress. Here, we investigate mitochondrial activity and explore stress-response pathways in preeclamptic placentas. We demonstrate activation of noncanonical mitochondrial unfolded protein response (UPRmt) pathways associated with reduced CLPP, a key protease in UPRmt signalling, that compromises mitochondrial respiration. The changes can be recapitulated in trophoblast cells by hypoxia–reoxygenation. Either activation of UPRmt or knockdown of CLPP can sufficiently reduce mitochondrial respiration. Translation of CLPP is negatively regulated by the endoplasmic reticulum UPR pathway. Understanding mitochondrial stress provides new insights into the pathophysiology of early-onset preeclampsia. Preeclampsia (PE) is a dangerous complication of pregnancy, especially when it presents at <34 wk of gestation (PE < 34 wk). It is a major cause of maternal and fetal morbidity and mortality and also increases the risk of cardiometabolic diseases in later life for both mother and offspring. Placental oxidative stress induced by defective placentation sits at the epicenter of the pathophysiology. The placenta is susceptible to activation of the unfolded protein response (UPR), and we hypothesized this may affect mitochondrial function. We first examined mitochondrial respiration before investigating evidence of mitochondrial UPR (UPRmt) in placentas of PE < 34 wk patients. Reduced placental oxidative phosphorylation (OXPHOS) capacity measured in situ was observed despite no change in protein or mRNA levels of electron transport chain complexes. These results were fully recapitulated by subjecting trophoblast cells to repetitive hypoxia–reoxygenation and were associated with activation of a noncanonical UPRmt pathway; the quality-control protease CLPP, central to UPRmt signal transduction, was reduced, while the cochaperone, TID1, was increased. Transcriptional factor ATF5, which regulates expression of key UPRmt genes including HSP60 and GRP75, showed no nuclear translocation. Induction of the UPRmt with methacycline reduced OXPHOS capacity, while silencing CLPP was sufficient to reduce OXPHOS capacity, membrane potential, and promoted mitochondrial fission. CLPP was negatively regulated by the PERK-eIF2α arm of the endoplasmic reticulum UPR pathway, independent of ATF4. Similar changes in the UPRmt pathway were observed in placentas from PE < 34 wk patients. Our results identify UPRmt as a therapeutic target for restoration of placental function in early-onset preeclampsia.
Collapse
|
198
|
Ghosh JC, Seo JH, Agarwal E, Wang Y, Kossenkov AV, Tang HY, Speicher DW, Altieri DC. Akt phosphorylation of mitochondrial Lonp1 protease enables oxidative metabolism and advanced tumor traits. Oncogene 2019; 38:6926-6939. [PMID: 31406245 PMCID: PMC6814529 DOI: 10.1038/s41388-019-0939-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/16/2019] [Accepted: 05/20/2019] [Indexed: 12/25/2022]
Abstract
Tumor mitochondria have heightened protein folding quality control, but the regulators of this process and how they impact cancer traits are not completely understood. Here we show that the ATP-directed mitochondrial protease, LonP1 is upregulated by stress conditions, including hypoxia, in tumor, but not normal cells. In mitochondria, LonP1 is phosphorylated by Akt on Ser173 and Ser181, enhancing its protease activity. Interference with this pathway induces accumulation of misfolded subunits of electron transport chain complex II and complex V, resulting in impaired oxidative bioenergetics and heightened ROS production. Functionally, this suppresses mitochondrial trafficking to the cortical cytoskeleton, shuts off tumor cell migration and invasion, and inhibits primary and metastatic tumor growth, in vivo. These data identify LonP1 as a key effector of mitochondrial reprogramming in cancer and potential therapeutic target.
Collapse
Affiliation(s)
- Jagadish C Ghosh
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Jae Ho Seo
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Ekta Agarwal
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Yuan Wang
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Hsin-Yao Tang
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - David W Speicher
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA.,Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, PA, 19104, USA.,Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, Philadelphia, PA, USA. .,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, 19104, USA.
| |
Collapse
|
199
|
Kreitz J, Schönfeld C, Seibert M, Stolp V, Alshamleh I, Oellerich T, Steffen B, Schwalbe H, Schnütgen F, Kurrle N, Serve H. Metabolic Plasticity of Acute Myeloid Leukemia. Cells 2019; 8:E805. [PMID: 31370337 PMCID: PMC6721808 DOI: 10.3390/cells8080805] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common and life-threatening leukemias. A highly diverse and flexible metabolism contributes to the aggressiveness of the disease that is still difficult to treat. By using different sources of nutrients for energy and biomass supply, AML cells gain metabolic plasticity and rapidly outcompete normal hematopoietic cells. This review aims to decipher the diverse metabolic strategies and the underlying oncogenic and environmental changes that sustain continuous growth, mediate redox homeostasis and induce drug resistance in AML. We revisit Warburg's hypothesis and illustrate the role of glucose as a provider of cellular building blocks rather than as a supplier of the tricarboxylic acid (TCA) cycle for energy production. We discuss how the diversity of fuels for the TCA cycle, including glutamine and fatty acids, contributes to the metabolic plasticity of the disease and highlight the roles of amino acids and lipids in AML metabolism. Furthermore, we point out the potential of the different metabolic effectors to be used as novel therapeutic targets.
Collapse
Affiliation(s)
- Johanna Kreitz
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
| | - Christine Schönfeld
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
| | - Marcel Seibert
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
| | - Verena Stolp
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
| | - Islam Alshamleh
- Center for Biomolecular Magnetic Resonance, Institute of Organic Chemistry and Chemical Biology, Goethe-University, 60438 Frankfurt am Main, Germany
| | - Thomas Oellerich
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany
| | - Björn Steffen
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany
| | - Harald Schwalbe
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
- Center for Biomolecular Magnetic Resonance, Institute of Organic Chemistry and Chemical Biology, Goethe-University, 60438 Frankfurt am Main, Germany
| | - Frank Schnütgen
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany
| | - Nina Kurrle
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany.
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany.
| | - Hubert Serve
- Department of Medicine 2, Hematology/Oncology, Goethe University, 60590 Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) and DKFZ, 69120 Heidelberg, Germany.
- Frankfurt Cancer Institute (FCI), 60590 Frankfurt am Main, Germany.
| |
Collapse
|
200
|
Hosseini M, Rezvani HR, Aroua N, Bosc C, Farge T, Saland E, Guyonnet-Dupérat V, Zaghdoudi S, Jarrou L, Larrue C, Sabatier M, Mouchel PL, Gotanègre M, Piechaczyk M, Bossis G, Récher C, Sarry JE. Targeting Myeloperoxidase Disrupts Mitochondrial Redox Balance and Overcomes Cytarabine Resistance in Human Acute Myeloid Leukemia. Cancer Res 2019; 79:5191-5203. [PMID: 31358527 DOI: 10.1158/0008-5472.can-19-0515] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/29/2019] [Accepted: 07/19/2019] [Indexed: 11/16/2022]
Abstract
Chemotherapies alter cellular redox balance and reactive oxygen species (ROS) content. Recent studies have reported that chemoresistant cells have an increased oxidative state in hematologic malignancies. In this study, we demonstrated that chemoresistant acute myeloid leukemia (AML) cells had a lower level of mitochondrial and cytosolic ROS in response to cytarabine (AraC) and overexpressed myeloperoxidase (MPO), a heme protein that converts hydrogen peroxide to hypochlorous acid (HOCl), compared with sensitive AML cells. High MPO-expressing AML cells were less sensitive to AraC in vitro and in vivo. They also produced higher levels of HOCl and exhibited an increased rate of mitochondrial oxygen consumption when compared with low MPO-expressing AML cells. Targeting MPO expression or enzyme activity sensitized AML cells to AraC treatment by triggering oxidative damage and sustaining oxidative stress, particularly in high MPO-expressing AML cells. This sensitization stemmed from mitochondrial superoxide accumulation, which impaired oxidative phosphorylation and cellular energetic balance, driving apoptotic death and selective eradication of chemoresistant AML cells in vitro and in vivo. Altogether, this study uncovers a noncanonical function of MPO enzyme in maintaining redox balance and mitochondrial energetic metabolism, therefore affecting downstream pathways involved in AML chemoresistance. SIGNIFICANCE: These findings demonstrate the role of myeloperoxidase in the regulation of ROS levels and sensitivity of AML cells to cytarabine, an essential chemotherapeutic backbone in the therapy of AML.
Collapse
Affiliation(s)
- Mohsen Hosseini
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Hamid Reza Rezvani
- INSERM U1035, Bordeaux, France.,Université de Bordeaux, Bordeaux, France
| | - Nesrine Aroua
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Claudie Bosc
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | | | - Sonia Zaghdoudi
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Latifa Jarrou
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Clément Larrue
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Pierre Luc Mouchel
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France.,Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Mathilde Gotanègre
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Equipe Labellisée LIGUE, Montpellier, France
| | - Guillaume Bossis
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Equipe Labellisée LIGUE, Montpellier, France
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France.,University of Toulouse, Toulouse, France.,Service d'Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, UMR1037, Inserm, Equipe Labellisée LIGUE 2018, Toulouse, France. .,University of Toulouse, Toulouse, France
| |
Collapse
|