1
|
Li W, Tu J, Zheng J, Das A, Yan Q, Jiang X, Ding W, Bai X, Lai K, Yang S, Yang C, Zou J, Diwan AD, Zheng Z. Gut Microbiome and Metabolome Changes in Chronic Low Back Pain Patients With Vertebral Bone Marrow Lesions. JOR Spine 2025; 8:e70042. [PMID: 39877797 PMCID: PMC11772216 DOI: 10.1002/jsp2.70042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Background Chronic low back pain (LBP) is a significant global health concern, often linked to vertebral bone marrow lesions (BML), particularly fatty replacement (FR). This study aims to explore the relationship between the gut microbiome, serum metabolome, and FR in chronic LBP patients. Methods Serum metabolomic profiling and gut microbiome analysis were conducted in chronic LBP patients with and without FR (LBP + FR, n = 40; LBP, n = 40) and Healthy Controls (HC, n = 31). The study investigates alterations in branched-chain amino acids (BCAAs) levels and identifies key microbial species associated with BCAA metabolism. In vitro experiments elucidate the role of BCAAs in adipogenesis of bone marrow mesenchymal stem cells (BM-MSCs) via the SIRT4 pathway. Results Chronic LBP patients with FR exhibit depleted BCAA levels in their serum metabolome, along with alterations in the gut microbiome. Specific microbial species, including Ruminococcus gnavus, Roseburia hominis, and Lachnospiraceae bacterium 8 1 57FAA, are identified as influential in BCAA metabolism and BM-MSCs metabolism. In vitro experiments demonstrate the ability of BCAAs to induce BM-MSCs adipogenesis through SIRT4 pathway activation. Conclusion This study sheds light on the intricate relationship between the disturbed gut ecosystem, serum metabolites, and FR in chronic LBP. Dysbiosis in the gut microbiome may contribute to altered BCAA degradation, subsequently promoting BM-MSCs adipogenesis and FR. Understanding these interactions provides insights for targeted therapeutic strategies to mitigate chronic LBP associated with FR by restoring gut microbial balance and modulating serum metabolite profiles.
Collapse
Affiliation(s)
- Wentian Li
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Gulbali Institute, School of Agricultural, Environmental and Veterinary SciencesCharles Sturt UniversityWagga WaggaAustralia
| | - Ji Tu
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Nepean HospitalNepean Blue Mountains Local Health DistrictPenrithAustralia
| | - Jinjian Zheng
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Abhirup Das
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
| | - Qi Yan
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiaotao Jiang
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Medicine and HealthThe University of new South WalesSydneyAustralia
| | - Wenyuan Ding
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Hebei Joint International Research Centre for Spinal DiseasesCenter for Innovation & Translational Medicine, the First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Xupeng Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Charles Perkins Centre and School of Medical SciencesUniversity of SydneySydneyAustralia
| | - Kaitao Lai
- Northcott Neuroscience LaboratoryANZAC Research Institute, Concord HospitalSydneyAustralia
- Tissue Engineering and Microfluidics Laboratory (TE&M)Australian Institute for Bioengineering and Nanotechnology (AIBN), the University of QueenslandSt LuciaAustralia
| | - Sidong Yang
- Department of Spinal SurgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cao Yang
- Spine ServiceDepartment of Orthopaedic Surgery, St. George HospitalKogarahAustralia
| | - Jun Zou
- Department of Orthopedic SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ashish D. Diwan
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhaomin Zheng
- Spine Labs, St. George and Sutherland Clinical SchoolUniversity of new South WalesKogarahAustralia
- Department of Spine Surgery, the First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
2
|
Zhang B, Mullmann J, Ludewig AH, Fernandez IR, Bales TR, Weiss RS, Schroeder FC. Acylspermidines are conserved mitochondrial sirtuin-dependent metabolites. Nat Chem Biol 2024; 20:812-822. [PMID: 38167917 PMCID: PMC11715332 DOI: 10.1038/s41589-023-01511-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
Sirtuins are nicotinamide adenine dinucleotide (NAD+)-dependent protein lysine deacylases regulating metabolism and stress responses; however, characterization of the removed acyl groups and their downstream metabolic fates remains incomplete. Here we employed untargeted comparative metabolomics to reinvestigate mitochondrial sirtuin biochemistry. First, we identified N-glutarylspermidines as metabolites downstream of the mitochondrial sirtuin SIR-2.3 in Caenorhabditis elegans and demonstrated that SIR-2.3 functions as a lysine deglutarylase and that N-glutarylspermidines can be derived from O-glutaryl-ADP-ribose. Subsequent targeted analysis of C. elegans, mouse and human metabolomes revealed a chemically diverse range of N-acylspermidines, and formation of N-succinylspermidines and/or N-glutarylspermidines was observed downstream of mammalian mitochondrial sirtuin SIRT5 in two cell lines, consistent with annotated functions of SIRT5. Finally, N-glutarylspermidines were found to adversely affect C. elegans lifespan and mammalian cell proliferation. Our results indicate that N-acylspermidines are conserved metabolites downstream of mitochondrial sirtuins that facilitate annotation of sirtuin enzymatic activities in vivo and may contribute to sirtuin-dependent phenotypes.
Collapse
Affiliation(s)
- Bingsen Zhang
- Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - James Mullmann
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
- Department of Molecular Medicine, Cornell University, Ithaca, NY, USA
| | | | - Irma R Fernandez
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Tyler R Bales
- Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, Cornell University, Ithaca, NY, USA
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, NY, USA.
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
3
|
Tataridas-Pallas N, Aman Y, Williams R, Chapman H, Cheng KJ, Gomez-Paredes C, Bates GP, Labbadia J. Mitochondrial clearance and increased HSF-1 activity are coupled to promote longevity in fasted Caenorhabditis elegans. iScience 2024; 27:109834. [PMID: 38784016 PMCID: PMC11112483 DOI: 10.1016/j.isci.2024.109834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 03/27/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Fasting has emerged as a potent means of preserving tissue function with age in multiple model organisms. However, our understanding of the relationship between food removal and long-term health is incomplete. Here, we demonstrate that in the nematode worm Caenorhabditis elegans, a single period of early-life fasting is sufficient to selectively enhance HSF-1 activity, maintain proteostasis capacity and promote longevity without compromising fecundity. These effects persist even when food is returned, and are dependent on the mitochondrial sirtuin, SIR-2.2 and the H3K27me3 demethylase, JMJD-3.1. We find that increased HSF-1 activity upon fasting is associated with elevated SIR-2.2 levels, decreased mitochondrial copy number and reduced H3K27me3 levels at the promoters of HSF-1 target genes. Furthermore, consistent with our findings in worms, HSF-1 activity is also enhanced in muscle tissue from fasted mice, suggesting that the potentiation of HSF-1 is a conserved response to food withdrawal.
Collapse
Affiliation(s)
- Nikolaos Tataridas-Pallas
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Yahyah Aman
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Rhianna Williams
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Hannah Chapman
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Kevin J.H. Cheng
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| | - Casandra Gomez-Paredes
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Gillian P. Bates
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - John Labbadia
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, Division of Biosciences, University College London, London WC1E 6BT, UK
| |
Collapse
|
4
|
Slade L, Bollen SE, Bass JJ, Phillips BE, Smith K, Wilkinson DJ, Szewczyk NJ, Atherton PJ, Etheridge T. Bisphosphonates attenuate age-related muscle decline in Caenorhabditis elegans. J Cachexia Sarcopenia Muscle 2023; 14:2613-2622. [PMID: 37722921 PMCID: PMC10751425 DOI: 10.1002/jcsm.13335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/17/2023] [Accepted: 08/21/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Age-related muscle decline (sarcopenia) associates with numerous health risk factors and poor quality of life. Drugs that counter sarcopenia without harmful side effects are lacking, and repurposing existing pharmaceuticals could expedite realistic clinical options. Recent studies suggest bisphosphonates promote muscle health; however, the efficacy of bisphosphonates as an anti-sarcopenic therapy is currently unclear. METHODS Using Caenorhabditis elegans as a sarcopenia model, we treated animals with 100 nM, 1, 10, 100 and 500 μM zoledronic acid (ZA) and assessed lifespan and healthspan (movement rates) using a microfluidic chip device. The effects of ZA on sarcopenia were examined using GFP-tagged myofibres or mitochondria at days 0, 4 and 6 post-adulthood. Mechanisms of ZA-mediated healthspan extension were determined using combined ZA and targeted RNAi gene knockdown across the life-course. RESULTS We found 100 nM and 1 μM ZA increased lifespan (P < 0.001) and healthspan [954 ± 53 (100 nM) and 963 ± 48 (1 μM) vs. 834 ± 59% (untreated) population activity AUC, P < 0.05]. 10 μM ZA shortened lifespan (P < 0.0001) but not healthspan (758.9 ± 37 vs. 834 ± 59, P > 0.05), whereas 100 and 500 μM ZA were larval lethal. ZA (1 μM) significantly improved myofibrillar structure on days 4 and 6 post-adulthood (83 and 71% well-organized myofibres, respectively, vs. 56 and 34% controls, P < 0.0001) and increased well-networked mitochondria at day 6 (47 vs. 16% in controls, P < 0.01). Genes required for ZA-mediated healthspan extension included fdps-1/FDPS-1 (278 ± 9 vs. 894 ± 17% population activity AUC in knockdown + 1 μM ZA vs. untreated controls, respectively, P < 0.0001), daf-16/FOXO (680 ± 16 vs. 894 ± 17%, P < 0.01) and agxt-2/BAIBA (531 ± 23 vs. 552 ± 8%, P > 0.05). Life/healthspan was extended through knockdown of igdb-1/FNDC5 (635 ± 10 vs. 523 ± 10% population activity AUC in gene knockdown vs. untreated controls, P < 0.01) and sir-2.3/SIRT-4 (586 ± 10 vs. 523 ± 10%, P < 0.05), with no synergistic improvements in ZA co-treatment vs. knockdown alone [651 ± 12 vs. 635 ± 10% (igdb-1/FNDC5) and 583 ± 9 vs. 586 ± 10% (sir-2.3/SIRT-4), both P > 0.05]. Conversely, let-756/FGF21 and sir-2.2/SIRT-4 were dispensable for ZA-induced healthspan [630 ± 6 vs. 523 ± 10% population activity AUC in knockdown + 1 μM ZA vs. untreated controls, P < 0.01 (let-756/FGF21) and 568 ± 9 vs. 523 ± 10%, P < 0.05 (sir-2.2/SIRT-4)]. CONCLUSIONS Despite lacking an endoskeleton, ZA delays Caenorhabditis elegans sarcopenia, which translates to improved neuromuscular function across the life course. Bisphosphonates might, therefore, be an immediately exploitable anti-sarcopenia therapy.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical SchoolExeterUK
- Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Shelby E. Bollen
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | - Joseph J. Bass
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | - Bethan E. Phillips
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | - Kenneth Smith
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | - Daniel J. Wilkinson
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | - Nathaniel J. Szewczyk
- Ohio Musculoskeletal and Neurological InstituteHeritage College of Osteopathic MedicineAthensOHUSA
| | - Philip J. Atherton
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research (CMAR), Unit of Injury, Recovery and Inflammation Sciences (IRIS), School of MedicineUniversity of NottinghamDerbyUK
| | | |
Collapse
|
5
|
Protein import motor complex reacts to mitochondrial misfolding by reducing protein import and activating mitophagy. Nat Commun 2022; 13:5164. [PMID: 36056001 PMCID: PMC9440083 DOI: 10.1038/s41467-022-32564-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
Mitophagy is essential to maintain mitochondrial function and prevent diseases. It activates upon mitochondria depolarization, which causes PINK1 stabilization on the mitochondrial outer membrane. Strikingly, a number of conditions, including mitochondrial protein misfolding, can induce mitophagy without a loss in membrane potential. The underlying molecular details remain unclear. Here, we report that a loss of mitochondrial protein import, mediated by the pre-sequence translocase-associated motor complex PAM, is sufficient to induce mitophagy in polarized mitochondria. A genome-wide CRISPR/Cas9 screen for mitophagy inducers identifies components of the PAM complex. Protein import defects are able to induce mitophagy without a need for depolarization. Upon mitochondrial protein misfolding, PAM dissociates from the import machinery resulting in decreased protein import and mitophagy induction. Our findings extend the current mitophagy model to explain mitophagy induction upon conditions that do not affect membrane polarization, such as mitochondrial protein misfolding. Mitophagy activation is mediated by mitochondrial depolarization. Here, the authors show that mitochondrial protein misfolding can activate mitophagy in a depolarization-independent manner mediated by a protein import reduction.
Collapse
|
6
|
Sirtuins and Autophagy in Age-Associated Neurodegenerative Diseases: Lessons from the C. elegans Model. Int J Mol Sci 2021; 22:ijms222212263. [PMID: 34830158 PMCID: PMC8619060 DOI: 10.3390/ijms222212263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 11/17/2022] Open
Abstract
Age-associated neurodegenerative diseases are known to have "impaired protein clearance" as one of the key features causing their onset and progression. Hence, homeostasis is the key to maintaining balance throughout the cellular system as an organism ages. Any imbalance in the protein clearance machinery is responsible for accumulation of unwanted proteins, leading to pathological consequences-manifesting in neurodegeneration and associated debilitating outcomes. Multiple processes are involved in regulating this phenomenon; however, failure to regulate the autophagic machinery is a critical process that hampers the protein clearing pathway, leading to neurodegeneration. Another important and widely known component that plays a role in modulating neurodegeneration is a class of proteins called sirtuins. These are class III histone deacetylases (HDACs) that are known to regulate various vital processes such as longevity, genomic stability, transcription and DNA repair. These enzymes are also known to modulate neurodegeneration in an autophagy-dependent manner. Considering its genetic relevance and ease of studying disease-related endpoints in neurodegeneration, the model system Caenorhabditis elegans has been successfully employed in deciphering various functional outcomes related to critical protein molecules, cell death pathways and their association with ageing. This review summarizes the vital role of sirtuins and autophagy in ageing and neurodegeneration, in particular highlighting the knowledge obtained using the C. elegans model system.
Collapse
|
7
|
Zaganjor E, Yoon H, Spinelli JB, Nunn ER, Laurent G, Keskinidis P, Sivaloganathan S, Joshi S, Notarangelo G, Mulei S, Chvasta MT, Tucker SA, Kalafut K, van de Ven RAH, Clish CB, Haigis MC. SIRT4 is an early regulator of branched-chain amino acid catabolism that promotes adipogenesis. Cell Rep 2021; 36:109345. [PMID: 34260923 DOI: 10.1016/j.celrep.2021.109345] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/23/2020] [Accepted: 06/15/2021] [Indexed: 11/30/2022] Open
Abstract
Upon nutrient stimulation, pre-adipocytes undergo differentiation to transform into mature adipocytes capable of storing nutrients as fat. We profiled cellular metabolite consumption to identify early metabolic drivers of adipocyte differentiation. We find that adipocyte differentiation raises the uptake and consumption of numerous amino acids. In particular, branched-chain amino acid (BCAA) catabolism precedes and promotes peroxisome proliferator-activated receptor gamma (PPARγ), a key regulator of adipogenesis. In early adipogenesis, the mitochondrial sirtuin SIRT4 elevates BCAA catabolism through the activation of methylcrotonyl-coenzyme A (CoA) carboxylase (MCCC). MCCC supports leucine oxidation by catalyzing the carboxylation of 3-methylcrotonyl-CoA to 3-methylglutaconyl-CoA. Sirtuin 4 (SIRT4) expression is decreased in adipose tissue of numerous diabetic mouse models, and its expression is most correlated with BCAA enzymes, suggesting a potential role for SIRT4 in adipose pathology through the alteration of BCAA metabolism. In summary, this work provides a temporal analysis of adipocyte differentiation and uncovers early metabolic events that stimulate transcriptional reprogramming.
Collapse
Affiliation(s)
- Elma Zaganjor
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA.
| | - Haejin Yoon
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica B Spinelli
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth R Nunn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Gaëlle Laurent
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Paulina Keskinidis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Suganja Sivaloganathan
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Giulia Notarangelo
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Stacy Mulei
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mathew T Chvasta
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Sarah A Tucker
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Krystle Kalafut
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Robert A H van de Ven
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Chen YY, Zhang XN, Xu CZ, Zhou DH, Chen J, Liu ZX, Sun Y, Huang W, Qu LS. MCCC2 promotes HCC development by supporting leucine oncogenic function. Cancer Cell Int 2021; 21:22. [PMID: 33407468 PMCID: PMC7788835 DOI: 10.1186/s12935-020-01722-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/12/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022] Open
Abstract
Background The role of methylcrotonoyl-CoA carboxylase 2 (MCCC2) in the development of tumors is well-established, and the involvement of leucine in the liver is well-known. However, the role of MCCC2 and the correlation between MCCC2 and leucine in the progression of hepatocellular carcinoma (HCC) have not yet been reported. Methods In this study, the Gepia database was used to evaluate the prognostic value of MCCC2 in HCC. The expression and localization of MCCC2 in HCC cells were determined by western blot and immunofluorescence assays. Flow cytometry and CCK-8 and transwell assays were carried out to explore the effect of MCCC2 on cell proliferation, migration, and invasion. In addition, mass spectrometry analysis was used to predict the potential cell function of MCCC2 in HCC. Results We found that the expression of MCCC2 increased in HCC tissues and that high expression of MCCC2 could predict poor outcomes in HCC patients. Knockdown expression of MCCC2 in HCC cells could reduce cell proliferation, migration, and invasion ability in vitro and could inhibit HCC cell proliferation in vivo. Interestingly, we found that HCC cells transfected with MCCC2-sgRNA failed to respond to leucine deprivation. Meanwhile, leucine deprivation inhibited cell proliferation, migration, and invasion in HCC cells where MCCC2 was present rather than in cells where MCCC2 was absent. In addition, knockdown of MCCC2 significantly reduced the glycolysis markers, glucose consumption, lactate secretion, and acetyl-CoA level, which is a product of leucine metabolism. Furthermore, we found that MCCC2 promotes the activation of ERK. Profiling the MCCC2 binding proteins revealed that MCCC2-associated proteins are enriched in biological processes, such as protein metabolism, energy pathway, and metabolism in HCC cells. Conclusions Our findings revealed that MCCC2 plays a critical role in the development of HCC, and the leucine metabolism pathway might be a novel target in HCC treatment.
Collapse
Affiliation(s)
- Yu-Yan Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Nantong University, Affiliated Hospital of Nantong University, Nantong, China
| | - Xue-Ning Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Chen-Zhou Xu
- Department of Gastroenterology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Dan-Hua Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Chen
- Research Center of Clinical Medicine, Nantong University, Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhao-Xiu Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ying Sun
- Blood Center of Jiangsu Province, Nanjing, China
| | - Wei Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.
| | - Li-Shuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
9
|
Betsinger CN, Cristea IM. Mitochondrial Function, Metabolic Regulation, and Human Disease Viewed through the Prism of Sirtuin 4 (SIRT4) Functions. J Proteome Res 2019; 18:1929-1938. [PMID: 30913880 DOI: 10.1021/acs.jproteome.9b00086] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
As cellular metabolic hubs, mitochondria are the main energy producers for the cell. These organelles host essential energy producing biochemical processes, including the TCA cycle, fatty acid oxidation, and oxidative phosphorylation. An accumulating body of literature has demonstrated that a majority of mitochondrial proteins are decorated with diverse posttranslational modifications (PTMs). Given the critical roles of these proteins in cellular metabolic pathways and response to environmental stress or pathogens, understanding the role of PTMs in regulating their functions has become an area of intense investigation. A major family of enzymes that regulate PTMs within the mitochondria are sirtuins (SIRTs). Albeit until recently the least understood sirtuin, SIRT4 has emerged as an enzyme capable of removing diverse PTMs from its substrates, thereby modulating their functions. SIRT4 was shown to have ADP-ribosyltransferase, deacetylase, lipoamidase, and deacylase enzymatic activities. As metabolic dysfunction is linked to human disease, SIRT4 levels and activities have been implicated in modulating susceptibility to hyperinsulinemia and diabetes, liver disease, cancer, neurodegeneration, heart disease, aging, and pathogenic infections. Therefore, SIRT4 has emerged as a possible candidate for targeted therapeutics. Here, we discuss the diverse enzymatic activities and substrates of SIRT4 and its roles in human health and disease.
Collapse
Affiliation(s)
- Cora N Betsinger
- Department of Molecular Biology , Princeton University , Princeton , New Jersey 08544 , United States
| | - Ileana M Cristea
- Department of Molecular Biology , Princeton University , Princeton , New Jersey 08544 , United States
| |
Collapse
|
10
|
Hada K, Hirota K, Inanobe A, Kako K, Miyata M, Araoi S, Matsumoto M, Ohta R, Arisawa M, Daitoku H, Hanada T, Fukamizu A. Tricarboxylic acid cycle activity suppresses acetylation of mitochondrial proteins during early embryonic development in Caenorhabditis elegans. J Biol Chem 2019; 294:3091-3099. [PMID: 30606736 DOI: 10.1074/jbc.ra118.004726] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 12/27/2018] [Indexed: 11/06/2022] Open
Abstract
The tricarboxylic acid (TCA) cycle (or citric acid cycle) is responsible for the complete oxidation of acetyl-CoA and formation of intermediates required for ATP production and other anabolic pathways, such as amino acid synthesis. Here, we uncovered an additional mechanism that may help explain the essential role of the TCA cycle in the early embryogenesis of Caenorhabditis elegans. We found that knockdown of citrate synthase (cts-1), the initial and rate-limiting enzyme of the TCA cycle, results in early embryonic arrest, but that this phenotype is not because of ATP and amino acid depletions. As a possible alternative mechanism explaining this developmental deficiency, we observed that cts-1 RNAi embryos had elevated levels of intracellular acetyl-CoA, the starting metabolite of the TCA cycle. Of note, we further discovered that these embryos exhibit hyperacetylation of mitochondrial proteins. We found that supplementation with acetylase-inhibiting polyamines, including spermidine and putrescine, counteracted the protein hyperacetylation and developmental arrest in the cts-1 RNAi embryos. Contrary to the hypothesis that spermidine acts as an acetyl sink for elevated acetyl-CoA, the levels of three forms of acetylspermidine, N 1-acetylspermidine, N 8-acetylspermidine, and N 1,N 8-diacetylspermidine, were not significantly increased in embryos treated with exogenous spermidine. Instead, we demonstrated that the mitochondrial deacetylase sirtuin 4 (encoded by the sir-2.2 gene) is required for spermidine's suppression of protein hyperacetylation and developmental arrest in the cts-1 RNAi embryos. Taken together, these results suggest the possibility that during early embryogenesis, acetyl-CoA consumption by the TCA cycle in C. elegans prevents protein hyperacetylation and thereby protects mitochondrial function.
Collapse
Affiliation(s)
- Kazumasa Hada
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan.,Department of Cell Biology, Faculty of Medicine, Oita University, 1-1 Hasama-machi Idaigaoka, Yufu, Oita, 879-5593, Japan
| | - Keiko Hirota
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan the.,Department of Hygiene and Public Health, School of Medicine, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Ai Inanobe
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Koichiro Kako
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan the
| | - Mai Miyata
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Sho Araoi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan
| | - Masaki Matsumoto
- Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Fukuoka, Fukuoka 812-8582, Japan
| | - Reiya Ohta
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan, and
| | - Mitsuhiro Arisawa
- Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamada-oka, Suita, Osaka 565-0871, Japan, and
| | - Hiroaki Daitoku
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Faculty of Medicine, Oita University, 1-1 Hasama-machi Idaigaoka, Yufu, Oita, 879-5593, Japan
| | - Akiyoshi Fukamizu
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8577, Japan, .,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8572, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
11
|
Sangaletti R, D’Amico M, Grant J, Della-Morte D, Bianchi L. Knock-out of a mitochondrial sirtuin protects neurons from degeneration in Caenorhabditis elegans. PLoS Genet 2017; 13:e1006965. [PMID: 28820880 PMCID: PMC5576752 DOI: 10.1371/journal.pgen.1006965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 08/30/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022] Open
Abstract
Sirtuins are NAD⁺-dependent deacetylases, lipoamidases, and ADP-ribosyltransferases that link cellular metabolism to multiple intracellular pathways that influence processes as diverse as cell survival, longevity, and cancer growth. Sirtuins influence the extent of neuronal death in stroke. However, different sirtuins appear to have opposite roles in neuronal protection. In Caenorhabditis elegans, we found that knock-out of mitochondrial sirtuin sir-2.3, homologous to mammalian SIRT4, is protective in both chemical ischemia and hyperactive channel induced necrosis. Furthermore, the protective effect of sir-2.3 knock-out is enhanced by block of glycolysis and eliminated by a null mutation in daf-16/FOXO transcription factor, supporting the involvement of the insulin/IGF pathway. However, data in Caenorhabditis elegans cell culture suggest that the effects of sir-2.3 knock-out act downstream of the DAF-2/IGF-1 receptor. Analysis of ROS in sir-2.3 knock-out reveals that ROS become elevated in this mutant under ischemic conditions in dietary deprivation (DD), but to a lesser extent than in wild type, suggesting more robust activation of a ROS scavenging system in this mutant in the absence of food. This work suggests a deleterious role of SIRT4 during ischemic processes in mammals that must be further investigated and reveals a novel pathway that can be targeted for the design of therapies aimed at protecting neurons from death in ischemic conditions.
Collapse
Affiliation(s)
- Rachele Sangaletti
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Massimo D’Amico
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Jeff Grant
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Department of Neurology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
- San Raffaele Roma Open University, Rome, Italy
| | - Laura Bianchi
- Department of Physiology and Biophysics, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
12
|
Kumar S, Lombard DB. For Certain, SIRT4 Activities! Trends Biochem Sci 2017; 42:499-501. [PMID: 28587732 DOI: 10.1016/j.tibs.2017.05.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 10/19/2022]
Abstract
Despite the fact that SIRT4 regulates important biological processes, its primary enzymatic activity has remained ambiguous. A recent study by Anderson, Huynh et al. has uncovered deacylase activities of SIRT4 towards newly described lysine modifications derived from reactive acyl-CoAs generated in leucine catabolism.
Collapse
Affiliation(s)
- Surinder Kumar
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - David B Lombard
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Institute of Gerontology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Anderson KA, Huynh FK, Fisher-Wellman K, Stuart JD, Peterson BS, Douros JD, Wagner GR, Thompson JW, Madsen AS, Green MF, Sivley RM, Ilkayeva OR, Stevens RD, Backos DS, Capra JA, Olsen CA, Campbell JE, Muoio DM, Grimsrud PA, Hirschey MD. SIRT4 Is a Lysine Deacylase that Controls Leucine Metabolism and Insulin Secretion. Cell Metab 2017; 25:838-855.e15. [PMID: 28380376 PMCID: PMC5444661 DOI: 10.1016/j.cmet.2017.03.003] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 09/26/2016] [Accepted: 03/06/2017] [Indexed: 01/17/2023]
Abstract
Sirtuins are NAD+-dependent protein deacylases that regulate several aspects of metabolism and aging. In contrast to the other mammalian sirtuins, the primary enzymatic activity of mitochondrial sirtuin 4 (SIRT4) and its overall role in metabolic control have remained enigmatic. Using a combination of phylogenetics, structural biology, and enzymology, we show that SIRT4 removes three acyl moieties from lysine residues: methylglutaryl (MG)-, hydroxymethylglutaryl (HMG)-, and 3-methylglutaconyl (MGc)-lysine. The metabolites leading to these post-translational modifications are intermediates in leucine oxidation, and we show a primary role for SIRT4 in controlling this pathway in mice. Furthermore, we find that dysregulated leucine metabolism in SIRT4KO mice leads to elevated basal and stimulated insulin secretion, which progressively develops into glucose intolerance and insulin resistance. These findings identify a robust enzymatic activity for SIRT4, uncover a mechanism controlling branched-chain amino acid flux, and position SIRT4 as a crucial player maintaining insulin secretion and glucose homeostasis during aging.
Collapse
Affiliation(s)
- Kristin A Anderson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Frank K Huynh
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Kelsey Fisher-Wellman
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - J Darren Stuart
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Brett S Peterson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Jonathan D Douros
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Gregory R Wagner
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - J Will Thompson
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Duke Proteomics and Metabolomics Shared Resource, Duke University Medical Center, Durham, NC 27710, USA
| | - Andreas S Madsen
- Center for Biopharmaceuticals and Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Michelle F Green
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - R Michael Sivley
- Department of Biological Sciences, Department of Biomedical Informatics, Vanderbilt Genetics Institute, Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Robert D Stevens
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Donald S Backos
- Computational Chemistry and Biology Core Facility, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - John A Capra
- Department of Biological Sciences, Department of Biomedical Informatics, Vanderbilt Genetics Institute, Center for Structural Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Christian A Olsen
- Center for Biopharmaceuticals and Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Regmi SG, Rolland SG. New Imaging Tools to Analyze Mitochondrial Morphology in Caenorhabditis elegans. Methods Mol Biol 2017; 1567:255-272. [PMID: 28276024 DOI: 10.1007/978-1-4939-6824-4_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mitochondria are highly dynamic organelles that constantly fuse and divide. This process is essential as several neurodegenerative diseases have been associated with defects in mitochondrial fusion or fission. Several tools have been developed over the years to visualize mitochondria in organisms such as Caenorhabditis elegans. Combining these tools with the powerful genetics of C. elegans has led to the discovery of new regulators of mitochondrial morphology. In this chapter, we present additional tools to further characterize mitochondrial morphology as well as regulators of mitochondrial morphology. Specifically, we introduce a photoactivatable mitoGFP (PAmitoGFP) that allows to investigate the connectivity of complex mitochondrial networks. In addition, we describe an immunostaining protocol that enables localization studies of these newly identified regulators of mitochondrial morphology.
Collapse
Affiliation(s)
- Saroj G Regmi
- LMU Biocenter, Department Biology II, Ludwig-Maximilians-University Munich, Grosshadernerstr. 2, Planegg-Martinsried, Munich, 82152, Germany.,Department of Genetics, Geisel School of Medicine at Dartmouth, 7400 Remsen, Hanover, NH, 03755, USA.,National Institute of Child Health and Human Development, NIH, 18 Center Drive, Bethesda, 20892, MD, USA
| | - Stéphane G Rolland
- LMU Biocenter, Department Biology II, Ludwig-Maximilians-University Munich, Grosshadernerstr. 2, Planegg-Martinsried, Munich, 82152, Germany.
| |
Collapse
|
15
|
Gaffney CJ, Shephard F, Chu J, Baillie DL, Rose A, Constantin-Teodosiu D, Greenhaff PL, Szewczyk NJ. Degenerin channel activation causes caspase-mediated protein degradation and mitochondrial dysfunction in adult C. elegans muscle. J Cachexia Sarcopenia Muscle 2016; 7:181-92. [PMID: 27493871 PMCID: PMC4864282 DOI: 10.1002/jcsm.12040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/17/2015] [Accepted: 04/09/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Declines in skeletal muscle structure and function are found in various clinical populations, but the intramuscular proteolytic pathways that govern declines in these individuals remain relatively poorly understood. The nematode Caenorhabditis elegans has been developed into a model for identifying and understanding these pathways. Recently, it was reported that UNC-105/degenerin channel activation produced muscle protein degradation via an unknown mechanism. METHODS Generation of transgenic and double mutant C. elegans, RNAi, and drug treatments were utilized to assess molecular events governing protein degradation. Western blots were used to measure protein content. Cationic dyes and adenosine triphosphate (ATP) production assays were utilized to measure mitochondrial function. RESULTS unc-105 gain-of-function mutants display aberrant muscle protein degradation and a movement defect; both are reduced in intragenic revertants and in let-2 mutants that gate the hyperactive UNC-105 channel. Degradation is not suppressed by interventions suppressing proteasome-mediated, autophagy-mediated, or calpain-mediated degradation nor by suppressors of degenerin-induced neurodegeneration. Protein degradation, but not the movement defect, is decreased by treatment with caspase inhibitors or RNAi against ced-3 or ced-4. Adult unc-105 muscles display a time-dependent fragmentation of the mitochondrial reticulum that is associated with impaired mitochondrial membrane potential and that correlates with decreased rates of maximal ATP production. Reduced levels of CED-4, which is sufficient to activate CED-3 in vitro, are observed in unc-105 mitochondrial isolations. CONCLUSIONS Constitutive cationic influx into muscle appears to cause caspase degradation of cytosolic proteins as the result of mitochondrial dysfunction, which may be relevant to ageing and sarcopenia.
Collapse
Affiliation(s)
- Christopher J Gaffney
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences University of Nottingham Nottingham NG7 2UH UK
| | - Freya Shephard
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences University of Nottingham Nottingham NG7 2UH UK
| | - Jeff Chu
- Department of Molecular Biology and Biochemistry Simon Fraser University Burnaby BCV5A 1S6 Canada; Department of Medical Genetics University of British Columbia Vancouver BCV6T 1Z4 Canada
| | - David L Baillie
- Department of Molecular Biology and Biochemistry Simon Fraser University Burnaby BC V5A 1S6 Canada
| | - Ann Rose
- Department of Medical Genetics University of British Columbia Vancouver BC V6T 1Z4 Canada
| | - Dumitru Constantin-Teodosiu
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences University of Nottingham Nottingham NG7 2UH UK
| | - Paul L Greenhaff
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences University of Nottingham Nottingham NG7 2UH UK
| | - Nathaniel J Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Faculty of Medicine and Health Sciences University of Nottingham Nottingham NG7 2UH UK
| |
Collapse
|
16
|
Mathias RA, Greco TM, Oberstein A, Budayeva HG, Chakrabarti R, Rowland EA, Kang Y, Shenk T, Cristea IM. Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell 2015; 159:1615-25. [PMID: 25525879 DOI: 10.1016/j.cell.2014.11.046] [Citation(s) in RCA: 326] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 11/12/2014] [Accepted: 11/21/2014] [Indexed: 12/20/2022]
Abstract
Sirtuins (SIRTs) are critical enzymes that govern genome regulation, metabolism, and aging. Despite conserved deacetylase domains, mitochondrial SIRT4 and SIRT5 have little to no deacetylase activity, and a robust catalytic activity for SIRT4 has been elusive. Here, we establish SIRT4 as a cellular lipoamidase that regulates the pyruvate dehydrogenase complex (PDH). Importantly, SIRT4 catalytic efficiency for lipoyl- and biotinyl-lysine modifications is superior to its deacetylation activity. PDH, which converts pyruvate to acetyl-CoA, has been known to be primarily regulated by phosphorylation of its E1 component. We determine that SIRT4 enzymatically hydrolyzes the lipoamide cofactors from the E2 component dihydrolipoyllysine acetyltransferase (DLAT), diminishing PDH activity. We demonstrate SIRT4-mediated regulation of DLAT lipoyl levels and PDH activity in cells and in vivo, in mouse liver. Furthermore, metabolic flux switching via glutamine stimulation induces SIRT4 lipoamidase activity to inhibit PDH, highlighting SIRT4 as a guardian of cellular metabolism.
Collapse
Affiliation(s)
- Rommel A Mathias
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA; Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Adam Oberstein
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Hanna G Budayeva
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Rumela Chakrabarti
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Elizabeth A Rowland
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Thomas Shenk
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
17
|
Jedrusik-Bode M. C. elegans sirtuin SIR-2.4 and its mammalian homolog SIRT6 in stress response. WORM 2014; 3:e29102. [PMID: 26442197 DOI: 10.4161/worm.29102] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 04/26/2014] [Accepted: 05/02/2014] [Indexed: 01/09/2023]
Abstract
Stress is a significant life event. The immediate response to stress is critical for survival. In organisms ranging from the unicellular Saccharomyces cerevisiae to protozoa (Trypanosoma brucei) and metazoan (such as Caenorhabditis elegans, Homo sapiens) stress response leads to the formation of cytoplasmic RNA-protein complexes referred to as stress granules (SGs). SGs regulate cell survival during stress by the sequestration of the signaling molecules implicated in apoptosis. They are a transient place of messenger ribonucleoproteins (mRNPs) remodeling for storage, degradation, or reinitiation of translation during stress and recovery from stress. Recently, we have identified chromatin factor, the sirtuin C. elegans SIR-2.4 variant and its mammalian homolog SIRT6 as a regulator of SGs formation. SIRT6 is highly conserved NAD(+)-dependent lysine deacetylase and ADP-ribosyltransferase impacting longevity, metabolism, and cancer. We observed that the cellular formation of SGs by SIRT6 or SIR-2.4 was linked with the cell viability or C. elegans survival and was dependent on SIRT6 enzymatic activity. Here, we discuss how SIR-2.4/SIRT6 influences SGs formation and stress response. We suggest possible mechanisms for such an unanticipated function of a chromatin regulatory factor SIRT6 in assembly of stress granules and cellular stress resistance.
Collapse
Affiliation(s)
- Monika Jedrusik-Bode
- Max Planck Institute for Biophysical Chemistry; Epigenetics in C elegans group; Göttingen, Germany
| |
Collapse
|