1
|
De Benedittis G, Latini A, Morgante C, Bonini C, Cela E, Kroegler B, Luciano A, Chiocchi M, Cavalli F, Ora J, Rogliani P, Novelli G, Ciccacci C, Chimenti MS, Conigliaro P, Borgiani P. Alteration of telomere length and mtDNA copy number in interstitial lung disease associated with rheumatoid arthritis. Autoimmunity 2025; 58:2473741. [PMID: 40035723 DOI: 10.1080/08916934.2025.2473741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/06/2025]
Abstract
Interstitial lung disease (ILD) is a common extra-articular manifestation of rheumatoid arthritis (RA). The inflammatory response in lung disease is characterized by severe oxidative stress, which enhances cellular senescence. Telomeric shortening and mitochondria dysregulation represent two hallmarks of cellular senescence. The maintenance of telomere length (TL) and mitochondrial DNA (mtDNA) copy number is preserved by many proteins, such as TERF1 and TFAM, respectively. Our aim was to evaluate the TL, the mtDNA copy number and the expression of two regulator gene factors in RA patients with (RA-ILD) and without lung involvement (RA-NILD). Eighty-five RA patients and 21 healthy subjects were enrolled. Relative TL, mtDNA copy number, and expression analysis of TERF1 and TFAM genes were measured using qPCR assay. All RA patients present a statistically significant telomere shortening; in particular, RA-ILD patients show shorter TL compared to both controls and RA-NILD. Patients with Usual Interstitial Pneumonia pattern show a more evident shortening of TL. Lastly, both RA-ILD and RA-NILD patients present a significant decrease in mtDNA copy number compared to controls. The analysis of regulatory genes showed an increase in TERF1 expression in RA patients compared to controls, also after stratification in the two subgroups, and a decrease in TFAM expression in RA patients compared to controls. These results show that the alteration of TL and mtDNA copy number in RA patients is more evident in the presence of ILD. The hypothesis is that, in these patients, oxidative stress could accelerate the shortening of telomeres and the decrease of mtDNA copy number.
Collapse
Affiliation(s)
- Giada De Benedittis
- Department of Biomedicine and Prevention, Section of Genetics, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Latini
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Chiara Morgante
- Department of Biomedicine and Prevention, Section of Genetics, University of Rome "Tor Vergata", Rome, Italy
| | - Chiara Bonini
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Eneida Cela
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Barbara Kroegler
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandra Luciano
- Department of Diagnostic Imaging and Interventional Radiology, University of Rome Tor Vergata, Rome, Italy
| | - Marcello Chiocchi
- Department of Diagnostic Imaging and Interventional Radiology, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Cavalli
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Division of Respiratory Medicine, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Section of Genetics, University of Rome "Tor Vergata", Rome, Italy
- Giovanni Lorenzini Medical Foundation, Milan, Italy
| | - Cinzia Ciccacci
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Borgiani
- Department of Biomedicine and Prevention, Section of Genetics, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
2
|
Xun Q, Yang Q, Wang W, Zhu G. GLCCI1 ameliorates mitochondrial dysfunction in allergic asthma mice via DYRK1A/FAM117B-dependent NRF2 activation. Cell Signal 2025; 134:111929. [PMID: 40490147 DOI: 10.1016/j.cellsig.2025.111929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/30/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
Allergic asthma significantly impacts individuals' quality of life and work. This study aimed to investigate the specific mechanisms by which GLCCI1 regulated mitochondrial dysfunction in allergic asthma mice. In an ovalbumin (OVA)-induced allergic asthma mouse model, mitochondrial dysfunction in airway epithelium was observed, characterized by reduced ATP production, decreased mtDNA copy number, ROS accumulation, and mitophagy activation (upregulated PINK1/OPTN). In vitro experiments confirmed that OVA stimulation impaired mitochondrial membrane potential, exacerbated oxidative stress, and reduced cell viability in bronchial epithelial cells (BECs). Moreover, GLCCI1 regulated DYRK1A/FAM117B and KEAP1/NRF2 axis while inhibiting NRF2 ubiquitination degradation. Mechanistically, GLCCI1 overexpression reversed OVA-induced mitochondrial dysfunction by activating NRF2 signaling pathway via enhancing DYRK1A/FAM117B. In allergic asthma mice, GLCCI1 overexpression improved airway hyperresponsiveness, reduced inflammatory infiltration, restored alveolar structure, and decreased IL-4/IL-5/IL-13 levels. In summary, GLCCI1 ameliorated mitochondrial dysfunction in allergic asthma mice via the DYRK1A/FAM117B/NRF2 pathway, offering a potential therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Qiufen Xun
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Qing Yang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Guofeng Zhu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| |
Collapse
|
3
|
Toraman E, Ceylan H. Investigation of the effect of tannic acid on doxorubicin-ınduced testicular damage and functions in a rat model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04238-0. [PMID: 40347279 DOI: 10.1007/s00210-025-04238-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
Reproductive and testicular toxicity is one of the most important side effects of doxorubicin (DOX), a chemotherapy drug. In our study, we investigated the ameliorative effect of tannic acid (TA), a polyphenol, against DOX-induced testicular and reproductive toxicity in rats. For this purpose, rats divided into four groups (n = 5 each). Rats were treated with DOX cumulatively 30 mg/kg (5 mg/kg by six equal injections) for 2 weeks to induce testicular toxicity. Rats were then treated daily with TA (50 mg/kg) and DOX + TA combination. After treatments, animals were decapitated and testicular tissues were removed. Then, to examine the effect of DOX and TA treatments on oxidative stress, changes in Sod, Cat, Gpx, Gst, and Gr specific activities and mRNA levels were determined. To determine the effects on inflammation, changes in the expression levels of Tnf-α, IL6, Foxo1, Foxo3, Cox2, and Inos genes were examined. In order to examine the therapeutic effect of TA on the spermatogenesis process, mRNA levels of Dazl, Amh, and Ddx4, which regulate reproductive functions, were determined. Additionally, changes in oxidative damage markers GSH, MDA, 8-OHdG, iNOS, and TNF-a and changes in mitochondrial DNA copy number were also investigated. The results showed that DOX treatment caused a decrease in the levels of oxidative stress and reproductive parameters and an increase in inflammation and DNA damage parameters. However, it was determined that oxidative stress, inflammation, and tissue damage decreased in testicular tissue after TA treatment. In addition, it was observed that TA also improved the expression levels of reproductive genes. When all the data were evaluated together, it was determined that TA administration has a therapeutic effect against the damage and toxicity caused by DOX.
Collapse
Affiliation(s)
- Emine Toraman
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, 25240, Erzurum, Turkey.
| | - Hamid Ceylan
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, 25240, Erzurum, Turkey
| |
Collapse
|
4
|
Abrahamsen ML, Christensen IB, Laizāne L, Ahmed HI, Buch-Larsen K, Marina D, Andersson M, Schwarz P, Dela F, Gillberg L. Transient increase in mitochondrial respiration in blood cells from breast cancer patients following chemo- and radiotherapy. Clin Exp Med 2025; 25:142. [PMID: 40338368 PMCID: PMC12062163 DOI: 10.1007/s10238-025-01665-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025]
Abstract
Mitochondrial respiration in peripheral blood mononuclear cells (PBMCs) has previously been shown to increase after chemo- and radiotherapy in early-stage breast cancer (BC) patients, but the persistence of the increase remains unknown. This study assessed whether changes in mitochondrial respiration and content in PBMCs from postmenopausal BC patients persist up to 1 year after treatment. Thirty-four early-stage BC patients were studied before, shortly after, and six- and twelve-months post-treatment along with 20 healthy controls. Mitochondrial respiration was measured using high-resolution respirometry of intact and permeabilized PBMCs. Mitochondrial content was estimated by quantifying mitochondrial DNA relative to nuclear DNA via qPCR. The mitochondrial respiratory capacity of intact and permeabilized PBMCs from BC patients significantly increased after adjuvant chemo- and radiotherapy (+ 33% and + 30% for the maximal capacity of the electron transport system, ETS), consistent with previous findings. Importantly, the respiratory capacity returned to pre-treatment levels six months after treatment completion in both intact and permeabilized cells (- 23% and - 26% for the ETS). Healthy controls exhibited similar mitochondrial respiration but had increased mitochondrial content (+ 20%) compared to BC patients before treatment. In summary, chemo- and radiotherapy transiently increased mitochondrial respiration in PBMCs, returning to baseline within six months after treatment completion. This temporary rise in oxygen demand may reflect immune system activation.
Collapse
Affiliation(s)
- Marie-Louise Abrahamsen
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Ida Bager Christensen
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Linda Laizāne
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
- Laboratory of Sports and Nutrition Research, Riga Stradiņš University, Riga, Latvia
| | - Haboon Ismail Ahmed
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | | | - Djordje Marina
- Department of Nephrology and Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | | | - Peter Schwarz
- Department of Nephrology and Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Dela
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
- Laboratory of Sports and Nutrition Research, Riga Stradiņš University, Riga, Latvia
| | - Linn Gillberg
- Xlab, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark.
| |
Collapse
|
5
|
Eswaran S, Mascarenhas R, Kabekkodu SP. The ester derivative Palmitoylcarnitine abrogates cervical cancer cell survival by enhancing lipotoxicity and mitochondrial dysfunction. Cell Commun Signal 2025; 23:213. [PMID: 40319292 PMCID: PMC12048960 DOI: 10.1186/s12964-025-02218-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND In cervical cancer (CC), Double C2 Like Domain Beta (DOC2B) functions as a metastatic suppressor. The present study aims to determine whether ectopic expression of DOC2B causes global metabolomic changes in extracellular vesicles (EVs) and corresponds with its tumor suppressive properties. METHODS Using a retroviral method, we first ectopically expressed DOC2B in SiHa cells, which do not normally express DOC2B. RESULTS We observed that ectopically expressed DOC2B significantly altered the global metabolite profile of EVs. Metabolomics identified significant enrichment of palmitoylcarnitine (PC) in EVs upon ectopic expression of DOC2B. We identified that SiHa and HeLa cells exhibited greater cytotoxicity to PC than gingival fibroblast, HaCaT, Cal27, and MCF7. PC treatment reduced the growth, proliferation, and migration of SiHa and HeLa cells, via increasing apoptosis and decreasing S-Phase cells. PC treatment resulted in morphological alterations, decreased length and number of filopodia, and expression of proteins related to cell cycle progression, proliferation, and the epithelial-to-mesenchymal transition. Further, PC treatment caused mitochondrial morphological changes, increased mitochondrial membrane potential, and decreased mtDNA content. The decreased GSH activity, glucose consumption rate, and lactate production upon PC treatment suggest that PC can induce metabolic reprogramming in CC cells. Increased oxidative stress, calcium overload, lipid droplet accumulation, mitochondrial lipotoxicity, and mitophagy suggest that PC can cause mitochondrial dysfunction. N-acetyl cysteine (NAC) treatment reversed the cytotoxic effect of PC, via decreasing lipid peroxidation rate and increasing GSH activity. PC treatment enhanced the cytotoxic effect of cisplatin in CC. CONCLUSION DOC2B restoration or the use of PC may be employed as a novel therapeutic approach for CC.
Collapse
Affiliation(s)
- Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Roshan Mascarenhas
- Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
- Newcastle University Medicine Malaysia (NUMed), 1, Jalan Sarjana 1, Kota Ilmu, Educity@Iskandar, Iskandar Puteri, Johor, 79200, Malaysia
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
6
|
Kizir D, Toraman E, Karaman M. Molecular Changes Associated with Inflammation and Reproduction in Cadmium-Induced Testicular Toxicity: Mitigating Effect of Lactobacillus plantarum. Biol Trace Elem Res 2025:10.1007/s12011-025-04601-5. [PMID: 40186081 DOI: 10.1007/s12011-025-04601-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Cadmium (Cd) is a dangerous heavy metal that causes toxicity in humans and animals. Various protective agents are being investigated to ameliorate the toxic effect of Cd. Lactic acid bacteria are promising as a protective agent because of their ability to remove heavy metals from contaminated products. The aim of this study was to evaluate the effects of Lactobacillus plantarum on biomarkers associated with reproduction and inflammation in the testes of rats exposed to Cd. For the application, rats were randomly divided into four groups: Control group, Cd group, L. plantarum group, and combined group (Cd + L. plantarum). At the end of 21 days of oral administration, testicular tissues of the rats were removed and mRNA expression levels of genes associated with reproduction (Dazl, Amh, and Ddx4) and inflammation (Tnf-α, Il-6, Cox-2, Inos, Foxo1, Foxo3, and Nfkb) were determined. On the other hand, the amounts of NOS2/iNOS, 8-ohdg, and Tnfα were analyzed in the tissues. The mtDNA copy number was also investigated. Cd caused an increase in the expression level of inflammation-related genes, except Nfkb, and a decrease in the expression level of reproduction-related genes. It also increased the levels of TNF-α, iNOS, and 8-OHdG biomarkers and mtDNA copy number. However, L. plantarum treatment did not cause a significant change in these parameters. Moreover, L. plantarum exposure in combination with Cd attenuated the observed Cd-mediated molecular changes in testicular tissue. In conclusion, the findings suggest that L. plantarum administration may be beneficial against Cd-induced testicular toxicity and may be considered as a biological agent in the development of protective strategies against environmental pollutants with further studies.
Collapse
Affiliation(s)
- Duygu Kizir
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, 25240, Turkey
| | - Emine Toraman
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, 25240, Turkey
| | - Melike Karaman
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Erzurum, 25240, Turkey.
| |
Collapse
|
7
|
Teerawongsuwan S, Wiangwised K, Ngampiyakul N, Wanikorn N, Boonnithipaisit P, Khiuhok P, Aekudompong P, Narkwichean A, Pongmayteegul S, Rungsiwiwut R. Relative mtDNA copy number in embryo spent culture medium is not a reliable biomarker of human embryo aneuploidy. REPRODUCTION AND FERTILITY 2025; 6:e250001. [PMID: 40243775 PMCID: PMC12060771 DOI: 10.1530/raf-25-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 03/08/2025] [Accepted: 04/17/2025] [Indexed: 04/18/2025] Open
Abstract
Graphical abstract Abstract Mitochondrial DNA (mtDNA) from embryonic cells is released into the spent culture medium (SCM) during cellular processes, providing a potential biomarker of embryo health. Analysing mtDNA levels in SCM enables a non-invasive evaluation of embryo quality and potential developmental abnormalities. In this retrospective study, we aimed to investigate the relationship between relative mtDNA copy number in embryo SCM and key factors, including embryo fragmentation, morphological quality and chromosomal abnormalities. Fertilised embryos produced through intracytoplasmic sperm injection were cultured to the blastocyst stage in an incubator. Embryo fragmentation was assessed on day 3 using the Istanbul criteria, while morphological grading was evaluated on day 5 using the Gardner criteria. On day 5, trophectoderm (TE) biopsies were performed for preimplantation genetic testing for aneuploidy, followed by embryo cryopreservation and collection of embryo SCM. The mtDNA was quantified using quantitative PCR. Statistical analyses using the Mann-Whitney U and Kruskal-Wallis tests (significance at P < 0.05) showed that relative mtDNA copy number did not significantly differ among embryos with fragmentation levels <10%, 10-25% and >25% (P > 0.05). For blastocyst grading, which evaluates the inner cell mass (ICM) and TE, no significant difference was observed in relative mtDNA copy number between grades B and C for ICM (P = 0.190) and TE (P = 0.289). Furthermore, a trend towards higher relative mtDNA levels was observed in aneuploid than in euploid embryos, although the difference was not statistically significant. Thus, relative mtDNA copy number in SCM may not accurately reflect embryo characteristics, such as fragmentation, morphological grading or chromosomal abnormalities. Lay summary This study examined whether the amount of mitochondrial DNA (mtDNA) in the fluid used to culture embryos in the laboratory could indicate embryo quality. We assessed various factors, including the appearance of the embryos, the presence of fragmented cells and the occurrence of chromosomal abnormalities. Fertilized eggs were cultured until they developed into blastocysts, and the amount of mtDNA in the culture fluid was measured using a machine that detects genetic material. The results revealed no clear association between mtDNA levels and embryo appearance or fragmentation. Although embryos with chromosomal abnormalities had slightly more mtDNA, the difference was not statistically significant. These findings suggest that mtDNA in the culture fluid may not be a reliable marker for assessing embryo quality or chromosomal status.
Collapse
Affiliation(s)
- Sasipat Teerawongsuwan
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
| | | | | | - Nitid Wanikorn
- Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | | | - Panyada Khiuhok
- Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | | | - Amarin Narkwichean
- Paragon Fertility Clinic, Bangkok, Thailand
- Department of Obstetrics and Gynecology, Faculty of Medicine, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Sirinun Pongmayteegul
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
- Research Unit in Stem Cell Innovation and Tissue Engineering, Srinakharinwirot University, Bangkok, Thailand
| | - Ruttachuk Rungsiwiwut
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, Thailand
- Research Unit in Stem Cell Innovation and Tissue Engineering, Srinakharinwirot University, Bangkok, Thailand
| |
Collapse
|
8
|
Xu J, Wakai M, Xiong K, Yang Y, Prabakaran A, Wu S, Ahrens D, Molina-Portela MDP, Ni M, Bai Y, Shavlakadze T, Glass DJ. The pro-inflammatory cytokine IL6 suppresses mitochondrial function via the gp130-JAK1/STAT1/3-HIF1α/ERRα axis. Cell Rep 2025; 44:115403. [PMID: 40056415 DOI: 10.1016/j.celrep.2025.115403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 03/10/2025] Open
Abstract
Chronic inflammation and a decline in mitochondrial function are hallmarks of aging. Here, we show that the two mechanisms may be linked. We found that interleukin-6 (IL6) suppresses mitochondrial function in settings where PGC1 (both PGC1α and PGC1β) expression is low. This suppression is mediated by the JAK1/STAT1/3 axis, which activates HIF1α through non-canonical mechanisms involving upregulation of HIF1A and ERRα transcription, and subsequent stabilization of the HIF1A protein by ERRα. HIF1α, in turn, inhibits ERRα, which is a master regulator of mitochondrial biogenesis, thus contributing to the inhibition of mitochondrial function. When expressed at higher levels, PGC1 rescues ERRα to boost baseline mitochondrial respiration, including under IL6-treated conditions. Our study suggests that inhibition of the IL6 signaling axis could be a potential treatment for those inflammatory settings where mitochondrial function is compromised.
Collapse
Affiliation(s)
- Jianing Xu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - Matthew Wakai
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Kun Xiong
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yanfeng Yang
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Adithya Prabakaran
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Sophia Wu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Diana Ahrens
- Research Flow Cytometry Core, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | | | - Min Ni
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yu Bai
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Tea Shavlakadze
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - David J Glass
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| |
Collapse
|
9
|
Pal S, Huttner D, Verma NC, Nemirovsky T, Ziv O, Sher N, Yivgi-Ohana N, Meller A. Amplification-Free Quantification of Endogenous Mitochondrial DNA Copy Number Using Solid-State Nanopores. ACS NANO 2025; 19:11390-11402. [PMID: 40082088 PMCID: PMC11948453 DOI: 10.1021/acsnano.5c00732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Mitochondrial DNA (mtDNA) quantification is crucial in understanding mitochondrial dysfunction, which is linked to a variety of diseases, including cancer and neurodegenerative disorders. Traditional methods often rely on amplification-based techniques, which can introduce bias and lack the precision needed for clinical diagnostics. Solid-state nanopores, an emerging biosensing platform, have the advantage of offering single-molecule and label-free approaches by enabling the direct counting of DNA molecules without amplification. The ion-current signatures obtained from each DNA molecule contain rich information on the molecules' lengths and origin. In this study, we present an amplification-free method for mtDNA quantification using solid-state nanopores and machine learning. Intriguingly, we find that native (unamplified) mtDNA translocations harbor structurally distinctive features that can be exploited to specifically detect and quantify mtDNA copies over the background of genomic DNA fragments. By combining selective degradation of linear genomic DNA (gDNA) via exonuclease V with a support vector machine (SVM)-based model, we isolate and quantify mtDNA directly from biological samples. We validate our method using plasmids or isolated mtDNAs by spiking in predetermined quantities. We then quantify endogenous mtDNAs in a cancer cell line and in blood cells and compare our results with qPCR-based quantification of the mtDNA/nuclear DNA ratios. To elucidate the source of the ion-current signatures from the native mtDNA molecules, we perform synchronous electro-optical sensing of mtDNAs during passage through the nanopore after NHS ester reaction with fluorophore compounds. Our results show correlated electro-optical events, indicating that the mtDNA is complexed with packaging proteins. Our assay is robust, with a high classification accuracy and is capable of detecting mtDNA at picomolar levels, making it suitable for low-abundance samples. This technique requires minimal sample preparation and eliminates the need for amplification or purification steps. The developed approach has significant potential for point-of-care applications, offering a low-cost and scalable solution for accurate mtDNA quantification in clinical settings.
Collapse
Affiliation(s)
- Sohini Pal
- Faculty
of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | - Diana Huttner
- Faculty
of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | - Navneet C. Verma
- Faculty
of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | - Talya Nemirovsky
- Faculty
of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
| | - Oren Ziv
- Minovia
Therapeutics Ltd., Tirat Carmel 3902603, Israel
| | - Noa Sher
- Minovia
Therapeutics Ltd., Tirat Carmel 3902603, Israel
| | | | - Amit Meller
- Faculty
of Biomedical Engineering, Technion -IIT, Haifa 3200003, Israel
- Russell
Berrie Nanotechnology Institute, Technion -IIT, Haifa 3200003, Israel
| |
Collapse
|
10
|
Seino R, Kubo H, Nishikubo K, Fukunaga H. Radiation-induced impacts on mitochondrial DNA and the transgenerational genomic instability. ENVIRONMENT INTERNATIONAL 2025; 196:109315. [PMID: 39914081 DOI: 10.1016/j.envint.2025.109315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/23/2025] [Accepted: 02/01/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Mitochondrial genomes are dynamically evolving and are shaped by somatic mutation and selection throughout the female germline. In this study, we investigated the radiation-induced impacts on mitochondrial DNA in vitro and in vivo, as well as the transgenerational inheritance. METHODS Human cervical cancer HeLa cells and telomerase-immortalized normal fibroblast BJ1-hTERT cells were exposed to X-rays at 0.5-8 Gy. Also, we exposed 8-week-old female C57BL/6N mice to a single whole-body dose of 2 Gy of X-rays and mated them with healthy males 1 day after irradiation. We extracted DNA from the irradiated cells, female mice, and the 2-week-old pups, then examined the mitochondrial DNA copy numbers (mtDNAcns) and radiation-induced damage by real-time quantitative PCR. RESULTS The mtDNAcn levels in cells increased and the intact copy ratios decreased 24 h after irradiation, resulting in the delayed shift of heteroplasmy. Also, the peripheral blood-derived mtDNAcn levels in irradiated females increased 1 day after irradiation and the intact copy rates decreased, supporting the results in vitro. Furthermore, whole blood-derived mtDNAcn levels decreased in 2-week-old pups born from irradiated mothers, indicating the mitochondrial genomic instability. DISCUSSION We found the delayed induction of mtDNA damage in human cancer and noncancerous cells after irradiation, as well as the same trend in mice. Furthermore, to our knowledge, this is first to demonstrate the hereditary effects of radiation on the regulation of mitochondrial genome. These provide novel insights into the significance of radiation protection and preventive medicine for not only pre-pregnancy females but also the next generation.
Collapse
Affiliation(s)
- Ryosuke Seino
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Haruka Kubo
- Department of Health Sciences, School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kai Nishikubo
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Hisanori Fukunaga
- Department of Biomedical Science and Engineering, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan; Center for Environmental and Health Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
11
|
Katoch S, Chhimwal J, Singh D, Kumar D, Patial V. Picrosides-rich fraction from Picrorhiza kurroa attenuates steatohepatitis in zebrafish and mice by modulating lipid metabolism and inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156368. [PMID: 39827774 DOI: 10.1016/j.phymed.2025.156368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) has become a serious public health concern with high global prevalence. The lack of safe and efficient treatment for the condition demands exploring new therapeutic solutions. PURPOSE In the present study, we investigated the protective efficacy of picrosides-rich fraction (PF) from Picrorhiza kurroa against steatohepatitis and revealed the molecular mechanism of action. METHODS PF was prepared and characterized using UPLC analysis. Initially, the efficacy of PF was studied on the zebrafish model of NASH. Further, a Methionine and Choline-Deficient (MCD) diet-induced NASH model in mice was employed to evaluate the hepatoprotective efficacy of PF by utilizing biochemical, histopathological and molecular studies. RESULTS The UPLC analysis revealed the presence of 29.11% and 29.86% picroside I and II in the PF, respectively. In the zebrafish model of NASH, PF treatment reduced the hepatic lipid accumulation and modulated the expressions of lipogenic, inflammatory, oxidative, and cellular stress genes. Further, in MCD diet-induced NASH in mice, PF treatment showed a significant improvement in body weights and serum liver injury markers. Reduced degenerative changes and fibrous tissue was observed in the PF-treated groups. The downregulated expression of Srebp1c, Cd36, Fas, Chrebp, Pparγ, and Hnf4α showed anti-lipogenic potential of PF treatment. NASH development followed oxidative stress, mitochondrial dysfunction, and inflammation in the liver of mice. However, PF treatment encouraged mitochondrial biogenesis by upregulating Pgc1α, Tfam, and Nrf2 expressions. The elevated levels of NFκB, TNFα, IL6, TGFβ, and αSMA were also restored by PF, advocating its anti-inflammatory and anti-fibrogenic effect. CONCLUSION The present study revealed that PF ameliorate the progression of NASH by increasing mitochondrial biogenesis and decreasing lipogenesis, hepatic inflammation, and fibrosis.
Collapse
Affiliation(s)
- Swati Katoch
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Dinesh Kumar
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, Himachal Pradesh 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
12
|
Trajano LADSN, Siqueira PB, Pinheiro D, Farias TG, Santos MSD, Pires BRB, Fonseca ADSD, Mencalha AL. Effects of photobiomodulation in mitochondrial quantity, biogenesis and mitophagy-associated genes in breast cancer cells. Lasers Med Sci 2025; 40:38. [PMID: 39849271 DOI: 10.1007/s10103-025-04287-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 01/05/2025] [Indexed: 01/25/2025]
Abstract
In this article, we aim to evaluate the effects of photobiomodulation on mitochondria quantity, biogenesis, and mitophagy-associated genes in breast cancer (BC) cells. Both models were irradiated with a low-power infrared laser (880 nm, 150 mW) and amber LED (617 nm, 1500 mW), alone or simultaneously. We evaluated the mRNA expression of PINK1 and PGC-1α genes, and the mitochondrial number was assessed based on the ratio of mitochondrial DNA/genomic DNA (mtDNA/gDNA). No significant difference was observed in the mtDNA/gDNA ratio comparing the low-power infrared laser (LPIL) and LED-irradiated groups to their control counterparts. Similarly, no difference was observed in the mRNA levels of PINK1 and PGC-1α of the irradiated group with an LPIL and LED alone or in combination. In conclusion, PBM with LPIL and LED did not alter the mtDNA/gDNA ratio nor modulate the mRNA levels of the genes related to mitophagy and biogenesis in BC cells.
Collapse
Affiliation(s)
- Larissa Alexsandra da Silva Neto Trajano
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil.
- Pró-Reitoria de Pesquisa e pós-graduação, Mestrado Profissional em Ciências Aplicadas em Saúde, Universidade de Vassouras, Avenida Expedicionário Oswaldo de Almeida Ramos, 280, Vassouras, Rio de Janeiro, 27700000, Brazil.
| | - Priscyanne Barreto Siqueira
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Daphne Pinheiro
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Thayssa Gomes Farias
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Márcia Soares Dos Santos
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Bruno Ricardo Barreto Pires
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| | - Adenilson de Souza da Fonseca
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
- Departamento de Ciências Fisiológicas, Instituto Biomédico, niversidade Federal do Estado do Rio de Janeiro, Rua Frei Caneca, 94, 20211040, Rio de Janeiro, Brazil
| | - Andre Luiz Mencalha
- Departamento de Biofísica e Biometria Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Avenida 28 de Setembro, 87, fundos, Vila Isabel, Rio de Janeiro, 20551030, Brazil
| |
Collapse
|
13
|
Arya D, Pawar P, Gajbhiye R, Tandon D, Kothari P, Goankar R, Singh D. Status of sperm mitochondrial functions and DNA methylation in infertile men with clinical varicocele before and after treatment. Mol Cell Endocrinol 2025; 595:112393. [PMID: 39481748 DOI: 10.1016/j.mce.2024.112393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Varicocele has been associated with reduced male fertility potential. Treatment modalities for varicocele improve semen parameters, yet more than 50% of cases remain infertile. Varicocele-induced heat and hypoxia stress may affect sperm mitochondrial functions, possibly leading to aberrant epigenetic modifications. This study includes 30 fertile men and 40 infertile men with clinical varicocele. The effect of varicocele treatment (antioxidant supplementation and or varicocelectomy) was evaluated after 3 months of treatment. Mitochondrial membrane potential (MMP) and intracellular reactive oxygen species (iROS) were measured by flow cytometry using JC-1 and DCFDA, respectively. mtDNA copy number and deletions were determined by PCR. DNA methylation was analysed by pyrosequencing. Present investigations suggest that infertile men with varicocele have abnormal semen parameters; significantly low MMP, high iROS, and high mtDNA copy number. Semen parameters were improved in a subset of men of both the treatment modalities; however, it was noted that varicocelectomy helped better in improving sperm parameters compared to antioxidant treatment. Both treatment modalities helped in reducing iROS and mtDNA copy number significantly; however, they were noneffective in improving MMP. Altered DNA methylation at mitochondria D loop and mitochondrial structure and function genes UQCRC2, MIC60, TOM22, and LETM1 (promoter region) were observed in varicocele group. The DNA methylation levels were restored after varicocele treatment; however, the restoration was not consistent at all CpG sites. Both the treatment modalities helped in restoring the altered DNA methylation levels of mitochondrial genes but the restoration is nonhomogeneous across the studied CpG sites.
Collapse
Affiliation(s)
- Deepshikha Arya
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, 400012, India
| | - Prakash Pawar
- Lokmanya Tilak Municipal General Hospital, Sion, Mumbai, 400022, India
| | - Rahul Gajbhiye
- Clinical Research Laboratory, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, 400012, India
| | - Deepti Tandon
- Department of Clinical Research, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, 400012, India
| | - Priyank Kothari
- Topiwala National Medical College and Bai Yamunabai Laxman Nair Charitable Hospital, Mumbai, 400008, India
| | - Reshma Goankar
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, 400012, India
| | - Dipty Singh
- Department of Neuroendocrinology, ICMR-National Institute for Research in Reproductive and Child Health, Parel, Mumbai, 400012, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
14
|
Sagliocchi S, Schiano E, Acampora L, Iannuzzo F, Cicatiello AG, Miro C, Nappi A, Restolfer F, Stornaiuolo M, Zarrilli S, Guerra F, Tenore GC, Dentice M, Novellino E. AbaComplex Enhances Mitochondrial Biogenesis and Adipose Tissue Browning: Implications for Obesity and Glucose Regulation. Foods 2024; 14:48. [PMID: 39796338 PMCID: PMC11720057 DOI: 10.3390/foods14010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Adipose tissue, particularly white adipose tissue (WAT), plays a central role in energy storage and metabolic regulation. Excess WAT, especially visceral fat, is strongly linked to metabolic disorders such as obesity and type 2 diabetes. The browning of WAT, whereby white fat cells acquire characteristics of brown adipose tissue (BAT) with enhanced thermogenic capacity, represents a promising strategy to enhance metabolic health. In this study, we investigated the effects of chronic supplementation with an infusion based on lyophilized, thin nectarines rich in abscisic acid (ABA), named AbaComplex, on promoting browning of WAT and activating BAT in mice. Over 30 days, C57BL/6 mice were treated with the ABA-rich infusion, and various metabolic and molecular parameters were assessed. The results showed that the AbaComplex significantly increased the expression of browning markers, such as UCP1 and PGC1-α, in both visceral and subcutaneous WAT. Additionally, mitochondrial biogenesis and function were enhanced, evidenced by elevated mitochondrial DNA content and activity. The treatment also reduced the weight of WAT (both visceral and subcutaneous) and BAT and significantly improved glucose uptake in WAT via upregulation of GLUT4, suggesting enhanced insulin sensitivity. Overall, the pronounced browning effect in WAT underscores the potential of AbaComplex as a natural approach for combating obesity and improving metabolic health.
Collapse
Affiliation(s)
- Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Elisabetta Schiano
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Fortuna Iannuzzo
- Department of Pharmacy, University of Chieti-Pescara G. D’Annunzio, 66100 Chieti, Italy;
| | - Annunziata Gaetana Cicatiello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 59, 80131 Napoli, Italy; (M.S.); (G.C.T.)
| | - Stefano Zarrilli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Fabrizia Guerra
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 59, 80131 Napoli, Italy; (M.S.); (G.C.T.)
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Ettore Novellino
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
- Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
15
|
De Benedittis G, Latini A, Morgante C, Perricone C, Ceccarelli F, Novelli G, Novelli L, Ciccacci C, Borgiani P. The dysregulation of mitochondrial homeostasis-related genes could be involved in the decrease of mtDNA copy number in systemic lupus erythematosus patients. Immunol Res 2024; 72:1384-1392. [PMID: 39230799 PMCID: PMC11618193 DOI: 10.1007/s12026-024-09535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic multifactorial autoimmune disease. It is now widely demonstrated that oxidative stress (OS) plays an important role in the modulation of the pathogenesis of this disease. Mitochondrial DNA (mtDNA) is highly vulnerable to OS and it is known a decrease of mtDNA copy number in SLE patients. However, to date, it has not been investigated if this decrease is associated with a dysregulation of mitochondrial homeostasis genes. Our aim is to evaluate the amount of mtDNA copy number and the expression of the genes more involved in the mitochondrial homeostasis pathways, in peripheral blood mononuclear cells (PBMCs) of SLE patients and healthy controls. We analysed the amount of mtDNA in PBMCs of 72 SLE patients and 61 healthy controls by qPCR. Then, we investigated the expression variability of TFAM and SIRT1 (biogenesis), MFN1 and MFF (fusion/fission) and PRKN2 (mitophagy) genes in a subgroup of SLE patients and healthy controls. Interestingly, we have observed a highly significant decrease in mtDNA copies in SLE patients compared to healthy controls (P < 0.0001). In addition, we have shown that the expression levels of SIRT1, MFN1 and PRKN2 genes were significantly decreased in SLE patients with respect to healthy controls (P = 0.00001 for SIRT1, P = 0.0150 for MFN1 and P = 0.0009 for PRKN2). Lastly, we have reported a positive correlation between PRKN2 expression level and mtDNA copy number (P = 0.019, r = 0.475). In conclusion, our data confirm the impairment of mtDNA copy number in the disease and show for the first time a dysregulation of the mitochondrial homeostasis genes. These results could provide additional support to the important role of mitochondria in SLE development.
Collapse
Affiliation(s)
- Giada De Benedittis
- Department of Biomedicine and Prevention, Genetics Section, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Andrea Latini
- Department of Biomedicine and Prevention, Genetics Section, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Chiara Morgante
- Department of Biomedicine and Prevention, Genetics Section, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Department of Internal Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Genetics Section, University of Rome Tor Vergata, 00133, Rome, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, USA
| | - Lucia Novelli
- UniCamillus, Saint Camillus International University of Health Sciences, 00131, Rome, Italy
| | - Cinzia Ciccacci
- UniCamillus, Saint Camillus International University of Health Sciences, 00131, Rome, Italy
| | - Paola Borgiani
- Department of Biomedicine and Prevention, Genetics Section, University of Rome Tor Vergata, 00133, Rome, Italy
| |
Collapse
|
16
|
Lancaster M, Hafen P, Law A, Matias C, Meyer T, Fischer K, Miller M, Hao C, Gillespie P, McKinzie D, Brault J, Graham B. Sucla2 Knock-Out in Skeletal Muscle Yields Mouse Model of Mitochondrial Myopathy With Muscle Type-Specific Phenotypes. J Cachexia Sarcopenia Muscle 2024; 15:2729-2742. [PMID: 39482887 PMCID: PMC11634519 DOI: 10.1002/jcsm.13617] [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: 10/23/2023] [Revised: 08/14/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Pathogenic variants in subunits of succinyl-CoA synthetase (SCS) are associated with mitochondrial encephalomyopathy in humans. SCS catalyses the conversion of succinyl-CoA to succinate coupled with substrate-level phosphorylation of either ADP or GDP in the TCA cycle. This report presents a muscle-specific conditional knock-out (KO) mouse model of Sucla2, the ADP-specific beta subunit of SCS, generating a novel in vivo model of mitochondrial myopathy. METHODS The mouse model was generated using the Cre-Lox system, with the human skeletal actin (HSA) promoter driving Cre-recombination of a CRISPR-Cas9-generated Sucla2 floxed allele within skeletal muscle. Inactivation of Sucla2 was validated using RT-qPCR and western blot, and both enzyme activity and serum metabolites were quantified by mass spectrometry. To characterize the model in vivo, whole-body phenotyping was conducted, with mice undergoing a panel of strength and locomotor behavioural assays. Additionally, ex vivo contractility experiments were performed on the soleus (SOL) and extensor digitorum longus (EDL) muscles. SOL and EDL cryosections were also subject to imaging analyses to assess muscle fibre-specific phenotypes. RESULTS Molecular validation confirmed 68% reduction of Sucla2 transcript within the mutant skeletal muscle (p < 0.001) and 95% functionally reduced SUCLA2 protein (p < 0.0001). By 3 weeks of age, Sucla2 KO mice were 44% the size of controls by body weight (p < 0.0001). Mutant mice also exhibited 34%-40% reduced grip strength (p < 0.01) and reduced spontaneous exercise, spending about 88% less cumulative time on a running wheel (p < 0.0001). Contractile function was also perturbed in a muscle-specific manner; although no genotype-specific deficiencies were seen in EDL function, SUCLA2-deficient SOL muscles generated 40% less specific tetanic force (p < 0.0001), alongside slower contraction and relaxation rates (p < 0.001). Similarly, a SOL-specific threefold increase in mitochondria (p < 0.0001) was observed, with qualitatively increased staining for both COX and SDH, and the proportion of Type 1 myosin heavy chain expressing fibres within the SOL was nearly doubled (95% increase, p < 0.0001) in the Sucla2 KO mice compared with that in controls. CONCLUSIONS SUCLA2 loss within murine skeletal muscle yields a model of SCS-deficient mitochondrial myopathy with reduced body weight, muscle weakness and exercise intolerance. Physiological and morphological analyses of hindlimb muscles showed remarkable differences in ex vivo function and cellular consequences between the EDL and SOL muscles, with SOL muscles significantly more impacted by Sucla2 inactivation. This novel model will provide an invaluable tool for investigations of muscle-specific and fibre type-specific pathogenic mechanisms to better understand SCS-deficient myopathy.
Collapse
Affiliation(s)
- Makayla S. Lancaster
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Paul Hafen
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology, & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Division of ScienceIndiana University ColumbusColumbusIndianaUSA
| | - Andrew S. Law
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology, & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Catalina Matias
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology, & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Timothy Meyer
- Behavioral Phenotyping CoreIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kathryn Fischer
- Behavioral Phenotyping CoreIndiana University School of MedicineIndianapolisIndianaUSA
| | - Marcus Miller
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Chunhai Hao
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Patrick Gillespie
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - David McKinzie
- Behavioral Phenotyping CoreIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jeffrey J. Brault
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology, & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Brett H. Graham
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
17
|
Cinzori ME, Nicol M, Dewald AL, Goodrich JM, Zhou Z, Gardiner JC, Kerver JM, Dolinoy DC, Talge N, Strakovsky RS. Maternal mitochondrial DNA copy number and methylation as possible predictors of pregnancy outcomes in a Michigan pregnancy cohort. ENVIRONMENTAL EPIGENETICS 2024; 10:dvae021. [PMID: 39628676 PMCID: PMC11614404 DOI: 10.1093/eep/dvae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024]
Abstract
Little is understood about the roles of mitochondria in pregnancy-related adaptations. Therefore, we evaluated associations of maternal early-to-mid pregnancy mitochondrial DNA copy number (mtDNAcn) and mtDNA methylation with birth size and gestational length. Michigan women (n = 396) provided venous bloodspots at median 11 weeks gestation to quantify mtDNAcn marker NADH-ubiquinone oxidoreductase chain 1 (ND1) using real-time quantitative PCR and mtDNA methylation at several regions within four mitochondria-specific genes using pyrosequencing: MTTF (mitochondrially encoded tRNA phenylalanine), DLOOP (D-loop promoter region, heavy strand), CYTB (cytochrome b), and LDLR (D-loop promoter region, light strand). We abstracted gestational length and birthweight from birth certificates and calculated birthweight z-scores using published references. We used multivariable linear regression to evaluate associations of mtDNAcn and mtDNA methylation with birthweight and birthweight z-scores. Cox Proportional Hazards Models (PHMs) and quantile regression characterized associations of mitochondrial measures with gestational length. We also considered differences by fetal sex. Using linear regression and Cox PHMs, mtDNAcn was not associated with birth outcomes, whereas associations of mtDNA methylation with birth outcomes were inconsistent. However, using quantile regression, mtDNAcn was associated with shorter gestation in female newborns at the upper quantiles of gestational length, but with longer gestational length in males at the lower quantiles of gestational length. Maternal LDLR, DLOOP, and MTTF methylation was associated with longer gestational length in females at the upper quantiles and in males at lower gestational length quantiles. Maternal mtDNAcn and mtDNA methylation were associated with gestational length in babies born comparatively early or late, which could reflect adaptations in mitochondrial processes that regulate the length of gestation.
Collapse
Affiliation(s)
- Maria E Cinzori
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Megan Nicol
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States
| | - Alisa L Dewald
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zheng Zhou
- Department of Animal Science, Michigan State University, East Lansing, MI 48824, United States
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Jean M Kerver
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Nicole Talge
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI 48824, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, United States
| |
Collapse
|
18
|
Tsai JF, Yu FY, Liu BH. Citrinin disrupts microtubule assembly in cardiac cells: Impact on mitochondrial organization and function. CHEMOSPHERE 2024; 365:143352. [PMID: 39293683 DOI: 10.1016/j.chemosphere.2024.143352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/31/2024] [Accepted: 09/14/2024] [Indexed: 09/20/2024]
Abstract
Citrinin (CTN) is a mycotoxin commonly present in various foods and feeds worldwide, as well as dietary supplements in Asian countries, but the risks and cellular mechanisms associated with its cardiotoxicity remains unclear. In this study, RNA-seq analysis of CTN-treated H9c2 cardiac cells demonstrated significant perturbations in pathways related to microtubule cytoskeleton and mitochondrial network organization. CTN disrupted microtubule polymerization and downregulated mRNA levels of microtubule-assembling genes, Map2 and Tpx2, in H9c2 cardiac cells. Additionally, CTN interfered with the distribution of mitochondrial network along the microtubules, leading to the accumulation of dysfunctional mitochondria characterized by elevated superoxide levels and reduced membrane potential. This disruption also caused the buildup of lysosomes and ubiquitinated proteins, which hindered waste clearance in microtubule-disassembled H9c2 cells. Molecular docking analysis indicated that CTN could bind to the colchicine binding site on β-tubulin, thereby mimicking the microtubule-disrupting effect of colchicine. This study provides morphological, transcriptomic, and mechanistic evidence to elucidate the cardiotoxic mechanisms of CTN, which involve the dysregulated microtubule network, subsequent mitochondrial mislocalization, and impaired proteolysis of damaged proteins/organelles in cardiac cells. Our findings may enhance the fundamental understanding and facilitate future risk assessment of CTN.
Collapse
Affiliation(s)
- Jui-Feng Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Feng-Yih Yu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Biing-Hui Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
19
|
Lv Z, Wang Z, Hu J, Su H, Liu B, Lang Y, Yu Q, Liu Y, Fan X, Yang M, Shen N, Zhang D, Zhang X, Wang R. LncRNA PVT1 induces mitochondrial dysfunction of podocytes via TRIM56 in diabetic kidney disease. Cell Death Dis 2024; 15:697. [PMID: 39349450 PMCID: PMC11442824 DOI: 10.1038/s41419-024-07107-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
Mitochondrial dysfunction is a significant contributor to podocyte injury in diabetic kidney disease (DKD). While previous studies have shown that PVT1 might play a vital role in DKD, the precise molecular mechanisms are largely unknown. By analyzing the plasma and kidney tissues of DKD patients, we observed a significant upregulation of PVT1 expression, which exhibited a positive correlation with albumin/creatinine ratios and serum creatinine levels. Then, we generated mice with podocyte-specific deletion of PVT1 (Nphs2-Cre/Pvt1flox/flox) and confirmed that the deletion of PVT1 suppressed podocyte mitochondrial dysfunction and inflammation in addition to ameliorating diabetes-induced podocyte injury, glomerulopathy, and proteinuria. Subsequently, we cultured podocytes in vitro and observed that PVT1 expression was upregulated under hyperglycemic conditions. Mechanistically, we demonstrated that PVT1 was involved in mitochondrial dysfunction by interacting with TRIM56 post-transcriptionally to modulate the ubiquitination of AMPKα, leading to aberrant mitochondrial biogenesis and fission. Additionally, the release of mtDNA and mtROS from damaged mitochondria triggered inflammation in podocytes. Subsequently, we verified the important role of TRIM56 in vivo by constructing Nphs2-Cre/Trim56flox/flox mice, consistently with the results of Nphs2-Cre/Pvt1flox/flox mice. Together, our results revealed that upregulation of PVT1 could promote mitochondrial dysfunction and inflammation of podocyte by modulating TRIM56, highlighting a potential novel therapeutic target for DKD.
Collapse
Affiliation(s)
- Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Ziyang Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Jinxiu Hu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Hong Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bing Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Yating Lang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Qun Yu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Yue Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xiaoting Fan
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Meilin Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ning Shen
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Dongdong Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xia Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
20
|
Grandi FC, Astord S, Pezet S, Gidaja E, Mazzucchi S, Chapart M, Vasseur S, Mamchaoui K, Smeriglio P. Characterization of SMA type II skeletal muscle from treated patients shows OXPHOS deficiency and denervation. JCI Insight 2024; 9:e180992. [PMID: 39264856 PMCID: PMC11530132 DOI: 10.1172/jci.insight.180992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/10/2024] [Indexed: 09/14/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a recessive developmental disorder caused by the genetic loss or mutation of the gene SMN1 (survival of motor neuron 1). SMA is characterized by neuromuscular symptoms and muscle weakness. Several years ago, SMA treatment underwent a radical transformation, with the approval of 3 different SMN-dependent disease-modifying therapies. This includes 2 SMN2 splicing therapies - risdiplam and nusinersen. One main challenge for type II SMA patients treated with these drugs is ongoing muscle fatigue, limited mobility, and other skeletal problems. To date, few molecular studies have been conducted on SMA patient-derived tissues after treatment, limiting our understanding of what targets remain unchanged after the spinal cord-targeted therapies are applied. Therefore, we collected paravertebral muscle from 8 type II patients undergoing spinal surgery for scoliosis and 7 controls. We used RNA-seq to characterize their transcriptional profiles and correlate these molecular changes with muscle histology. Despite the limited cohort size and heterogeneity, we observed a consistent loss of oxidative phosphorylation (OXPHOS) machinery of the mitochondria, a decrease in mitochondrial DNA copy number, and a correlation between signals of cellular stress, denervation, and increased fibrosis. This work provides new putative targets for combination therapies for type II SMA.
Collapse
Affiliation(s)
- Fiorella Carla Grandi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Stéphanie Astord
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Sonia Pezet
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Elèna Gidaja
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Sabrina Mazzucchi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Maud Chapart
- Centre de Ressources Biologiques - Myobank-AFM de l’Institut de Myologie, Hôpital de la Pitié-Salpêtrière F - 75013 Paris, France
| | - Stéphane Vasseur
- Centre de Ressources Biologiques - Myobank-AFM de l’Institut de Myologie, Hôpital de la Pitié-Salpêtrière F - 75013 Paris, France
| | - Kamel Mamchaoui
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| | - Piera Smeriglio
- Sorbonne Université, INSERM, Institut de Myologie, Centre de recherche en Myologie F-75013 Paris, France
| |
Collapse
|
21
|
Giuga M, Ferrito V, Calogero GS, Traina A, Bonsignore M, Sprovieri M, Pappalardo AM. Differential Cellular Response to Mercury in Non-Farmed Fish Species Based on Mitochondrial DNA Copy Number Variation Analysis. BIOLOGY 2024; 13:691. [PMID: 39336118 PMCID: PMC11429374 DOI: 10.3390/biology13090691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/30/2024]
Abstract
Mercury (Hg) pro-oxidant role on biological systems and its biogeochemical cycle represent a serious threat due to its persistence in marine environment. As the mitochondrial genome is exposed to reactive oxygen species (ROS), the aim of the present study is the validation of the variation in the number of mitochondrial DNA copies (mtDNAcn) as biomarker of oxidative stress in aquatic environment. During summer 2021, three selected fish species (Mullus barbatus, Diplodus annularis and Pagellus erythrinus) were collected in Augusta Bay, one of the most Mediterranean contaminated areas remarkable by past Hg inputs, and in a control area, both in the south-east of Sicily. The relative mtDNAcn was evaluated by qPCR on specimens of each species from both sites, characterized respectively by higher and lower Hg bioaccumulation. M. barbatus and P. erythrinus collected in Augusta showed a dramatic mtDNAcn reduction compared to their control groups while D. annularis showed an incredible mtDNAcn rising suggesting a higher resilience of this species. These results align with the mitochondrial dynamics of fission and fusion triggered by environmental toxicants. In conclusion, we suggest the implementation of the mtDNAcn variation as a valid tool for the early warning stress-related impacts in aquatic system.
Collapse
Affiliation(s)
- Marta Giuga
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology "M. La Greca", University of Catania, Via Androne 81, 95124 Catania, Italy
- National Research Council of Italy, Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), Via De Marini 16, 16149 Genova, Italy
| | - Venera Ferrito
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology "M. La Greca", University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Giada Santa Calogero
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology "M. La Greca", University of Catania, Via Androne 81, 95124 Catania, Italy
| | - Anna Traina
- National Research Council of Italy, Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), Lungomare Cristoforo Colombo 452, 90149 Palermo, Italy
| | - Maria Bonsignore
- National Research Council of Italy, Institute of Anthropic Impacts and Sustainability in Marine Environment (CNR-IAS), Via del Mare, 91021 Campobello di Mazara, Italy
| | - Mario Sprovieri
- National Research Council of Italy, Institute of Marine Sciences (ISMAR-CNR), Tesa 104-Arsenale, Castello 2737/F, 30122 Venezia, Italy
| | - Anna Maria Pappalardo
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology "M. La Greca", University of Catania, Via Androne 81, 95124 Catania, Italy
| |
Collapse
|
22
|
Almeida EA, Mehndiratta M, Madhu SV, Kar R. Higher Body Mass Index is Associated with Increased Oxidative Stress in Patients of Type 2 Diabetes Mellitus. Indian J Endocrinol Metab 2024; 28:517-521. [PMID: 39676781 PMCID: PMC11642501 DOI: 10.4103/ijem.ijem_474_23] [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: 12/12/2023] [Revised: 01/30/2024] [Accepted: 02/28/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) is closely associated with the obesity; however, certain proportion of T2DM patients is non-obese or lean (BMI < 18.5 kg/m2). Obesity has long been associated with oxidative stress; however, there are no studies available documenting levels of oxidative stress in the lean patients of T2DM. Therefore, this study was done to compare the levels of makers of oxidative stress (TL, mtDNA, TAS) and their regulators (mRNA expression of TERT, Nrf2 and Nqo1) in lean and obese patients of T2DM. Methods 60 newly diagnosed patients (treatment naïve) of T2DM were recruited and divided into lean (BMI < 18.5 kg/m2) and obese (BMI > 25 kg/m2) groups. Relative telomere length (T/S) and mtDNA content were estimated via real-time PCR. Serum total antioxidant status (TAS) was measured using a commercially available kit. mRNA expression of TERT, Nrf2 and Nqo1 was measured by real-time PCR. Results Mean T/S and mtDNA content were lower in the obese group compared to the lean group (P = 0.16 and P = 0.06, respectively). Mean serum TAS levels were higher in obese group compared to the lean group (P = 0.001). mRNA expression of TERT and Nrf2 was increased in obese group compared to the lean group. mRNA expression of Nqo1 was similar in both the groups. Conclusion Obese patients of T2DM are exposed to a greater degree of OS compared to the lean patients of T2DM.
Collapse
Affiliation(s)
- Edelbert A. Almeida
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| | - Mohit Mehndiratta
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| | - Sri V. Madhu
- Department of Endocrinology, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| | - Rajarshi Kar
- Department of Biochemistry, University College of Medical Sciences and GTB Hospital, University of Delhi, Delhi, India
| |
Collapse
|
23
|
Li SJ, Wei JQ, Kang YY, Wang RQ, Rong WW, Zhao JJ, Deng QW, Gao PJ, Li XD, Wang JG. Natriuretic peptide receptor-C perturbs mitochondrial respiration in white adipose tissue. J Lipid Res 2024; 65:100623. [PMID: 39154732 PMCID: PMC11418126 DOI: 10.1016/j.jlr.2024.100623] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/15/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024] Open
Abstract
Natriuretic peptide receptor-C (NPR-C) is highly expressed in adipose tissues and regulates obesity-related diseases; however, the detailed mechanism remains unknown. In this research, we aimed to explore the potential role of NPR-C in cold exposure and high-fat/high-sugar (HF/HS) diet-induced metabolic changes, especially in regulating white adipose tissue (WAT) mitochondrial function. Our findings showed that NPR-C expression, especially in epididymal WAT (eWAT), was reduced after cold exposure. Global Npr3 (gene encoding NPR-C protein) deficiency led to reduced body weight, increased WAT browning, thermogenesis, and enhanced expression of genes related to mitochondrial biogenesis. RNA-sequencing of eWAT showed that Npr3 deficiency enhanced the expression of mitochondrial respiratory chain complex genes and promoted mitochondrial oxidative phosphorylation in response to cold exposure. In addition, Npr3 KO mice were able to resist obesity induced by HF/HS diet. Npr3 knockdown in stromal vascular fraction (SVF)-induced white adipocytes promoted the expression of proliferator-activated receptor gamma coactivator 1α (PGC1α), uncoupling protein one (UCP1), and mitochondrial respiratory chain complexes. Mechanistically, NPR-C inhibited cGMP and calcium signaling in an NPR-B-dependent manner but suppressed cAMP signaling in an NPR-B-independent manner. Moreover, Npr3 knockdown induced browning via AKT and p38 pathway activation, which were attenuated by Npr2 knockdown. Importantly, treatment with the NPR-C-specific antagonist, AP-811, decreased WAT mass and increased PGC-1α, UCP1, and mitochondrial complex expression. Our findings reveal that NPR-C deficiency enhances mitochondrial function and energy expenditure in white adipose tissue, contributing to improved metabolic health and resistance to obesity.
Collapse
Affiliation(s)
- Shi-Jin Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Jin-Qiu Wei
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Yuan Kang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Qi Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wu-Wei Rong
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Jia Zhao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian-Wan Deng
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping-Jin Gao
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Dong Li
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ji-Guang Wang
- Department of Cardiovascular Medicine, Department of Hypertension, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Frances A, Lumeau A, Bery N, Gayral M, Stuani L, Sorbara M, Saland E, Pagan D, Hanoun N, Torrisani J, Lemarié A, Portais JC, Buscail L, Dusetti N, Sarry JE, Cordelier P. Cytidine deaminase-dependent mitochondrial biogenesis as a potential vulnerability in pancreatic cancer cells. Commun Biol 2024; 7:1065. [PMID: 39215188 PMCID: PMC11364846 DOI: 10.1038/s42003-024-06760-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Cytidine deaminase (CDA) converts cytidine and deoxycytidine into uridine and deoxyuridine as part of the pyrimidine salvage pathway. Elevated levels of CDA are found in pancreatic tumors and associated with chemoresistance. Recent evidence suggests that CDA has additional functions in cancer cell biology. In this work, we uncover a novel role of CDA in pancreatic cancer cell metabolism. CDA silencing impairs mitochondrial metabolite production, respiration, and ATP production in pancreatic cancer cells, leading to a so-called Pasteur effect metabolic shift towards glycolysis. Conversely, we find that CDA expression promotes mitochondrial biogenesis and oxidative phosphorylation, independently of CDA deaminase activity. Furthermore, we observe that patient primary cells overexpressing CDA are more sensitive to mitochondria-targeting drugs. Collectively, this work shows that CDA plays a non-canonical role in pancreatic cancer biology by promoting mitochondrial function, which could be translated into novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Audrey Frances
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Audrey Lumeau
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Nicolas Bery
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Marion Gayral
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Lucille Stuani
- Team METAML-METabolism and Drug Resistance in Acute Myeloid Leukemia, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Equipe de Recherche Labélisée Ligue Nationale Contre le Cancer en 2023, Toulouse, France
- Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Toulouse, France
| | - Marie Sorbara
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Estelle Saland
- Team METAML-METabolism and Drug Resistance in Acute Myeloid Leukemia, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Equipe de Recherche Labélisée Ligue Nationale Contre le Cancer en 2023, Toulouse, France
- Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Toulouse, France
| | - Delphine Pagan
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Naïma Hanoun
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Jérôme Torrisani
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Anthony Lemarié
- Team Radiotherapy Optimising: From Molecular Signalling Pathways to Clinical Trials, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
| | - Jean-Charles Portais
- Institut RESTORE, UMR 1301 INSERM, 5070 CNRS, Université Paul Sabatier, Toulouse, France
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
- Toulouse Biotechnology Institute-INSA de Toulouse INSA/CNRS 5504-UMR INSA/INRA 792, Toulouse, France
| | - Louis Buscail
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Service de Gastroentérologie et de pancréatologir, CHU Rangueil, Université de Toulouse, Toulouse, France
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille, CRCM, Inserm, CNRS, Institut Paoli-Calmettes, Université Aix-Marseille, Marseille, France
| | - Jean-Emmanuel Sarry
- Team METAML-METabolism and Drug Resistance in Acute Myeloid Leukemia, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France
- Equipe de Recherche Labélisée Ligue Nationale Contre le Cancer en 2023, Toulouse, France
- Laboratoire d'Excellence Toulouse Cancer (TOUCAN), Toulouse, France
| | - Pierre Cordelier
- Team Therapeutic Innovation in Pancreatic Cancer, Centre de Recherches en Cancérologie de Toulouse, CRCT, Université de Toulouse, Inserm, CNRS, Toulouse, France.
| |
Collapse
|
25
|
Tohme C, Haykal T, Yang R, Austin TJ, Loughran P, Geller DA, Simmons RL, Tohme S, Yazdani HO. ZLN005, a PGC-1α Activator, Protects the Liver against Ischemia-Reperfusion Injury and the Progression of Hepatic Metastases. Cells 2024; 13:1448. [PMID: 39273020 PMCID: PMC11393917 DOI: 10.3390/cells13171448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Exercise can promote sustainable protection against cold and warm liver ischemia-reperfusion injury (IRI) and tumor metastases. We have shown that this protection is by the induction of hepatic mitochondrial biogenesis pathway. In this study, we hypothesize that ZLN005, a PGC-1α activator, can be utilized as an alternative therapeutic strategy. METHODS Eight-week-old mice were pretreated with ZLN005 and subjected to liver warm IRI. To establish a liver metastatic model, MC38 cancer cells (1 × 106) were injected into the spleen, followed by splenectomy and liver IRI. RESULTS ZLN005-pretreated mice showed a significant decrease in IRI-induced tissue injury as measured by serum ALT/AST/LDH levels and tissue necrosis. ZLN005 pretreatment decreased ROS generation and cell apoptosis at the site of injury, with a significant decrease in serum pro-inflammatory cytokines, innate immune cells infiltration, and intrahepatic neutrophil extracellular trap (NET) formation. Moreover, mitochondrial mass was significantly upregulated in hepatocytes and maintained after IRI. This was confirmed in murine and human hepatocytes treated with ZLN005 in vitro under normoxic and hypoxic conditions. Additionally, ZLN005 preconditioning significantly attenuated tumor burden and increased the percentage of intratumoral cytotoxic T cells. CONCLUSIONS Our study highlights the effective protection of ZLN005 pretreatment as a therapeutic alternative in terms of acute liver injury and tumor metastases.
Collapse
Affiliation(s)
- Celine Tohme
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Tony Haykal
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Ruiqi Yang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Taylor J. Austin
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
- Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - David A. Geller
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Richard L. Simmons
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Samer Tohme
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| | - Hamza O. Yazdani
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.T.); (T.H.); (R.Y.); (T.J.A.); (P.L.); (D.A.G.); (R.L.S.)
| |
Collapse
|
26
|
Chang-Chien J, Huang JL, Tsai HJ, Wang SL, Kuo ML, Yao TC. Vitamin D ameliorates particulate matter induced mitochondrial damages and calcium dyshomeostasis in BEAS-2B human bronchial epithelial cells. Respir Res 2024; 25:321. [PMID: 39174953 PMCID: PMC11342659 DOI: 10.1186/s12931-024-02951-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Mitochondria is prone to oxidative damage by endogenous and exogenous sources of free radicals, including particulate matter (PM). Given the role of mitochondria in inflammatory disorders, such as asthma and chronic obstructive pulmonary disease, we hypothesized that supplementation of vitamin D may play a protective role in PM-induced mitochondrial oxidative damages of human bronchial epithelial BEAS-2B cells. METHODS BEAS-2B cells were pretreated with 1,25(OH)2D3, an active form of vitamin D, for 1 h prior to 24-hour exposure to PM (SRM-1648a). Oxidative stress was measured by flow cytometry. Mitochondrial functions including mitochondrial membrane potential, ATP levels, and mitochondrial DNA copy number were analyzed. Additionally, mitochondrial ultrastructure was examined using transmission electron microscopy. Intracellular and mitochondrial calcium concentration changes were assessed using flow cytometry based on the expression of Fluo-4 AM and Rhod-2 AM, respectively. Pro-inflammatory cytokines, including IL-6 and MCP-1, were quantified using ELISA. The expression levels of antioxidants, including SOD1, SOD2, CAT, GSH, and NADPH, were determined. RESULTS Our findings first showed that 24-hour exposure to PM led to the overproduction of reactive oxygen species (ROS) derived from mitochondria. PM-induced mitochondrial oxidation resulted in intracellular calcium accumulation, particularly within mitochondria, and alterations in mitochondrial morphology and functions. These changes included loss of mitochondrial membrane integrity, disarrayed cristae, mitochondrial membrane depolarization, reduced ATP production, and increased mitochondrial DNA copy number. Consequently, PM-induced mitochondrial damage triggered the release of certain inflammatory cytokines, such as IL-6 and MCP-1. Similar to the actions of mitochondrial ROS inhibitor MitoTEMPO, 1,25(OH)2D3 conferred protective effects on mtDNA alterations, mitochondrial damages, calcium dyshomeostasis, thereby decreasing the release of certain inflammatory cytokines. We found that greater cellular level of 1,25(OH)2D3 upregulated the expression of enzymatic (SOD1, SOD2, and CAT) and non-enzymatic (GSH and NADPH) antioxidants to modulate cellular redox homeostasis. CONCLUSION Our study provides new evidence that 1,25(OH)2D3 acts as an antioxidant, enhancing BEAS-2B antioxidant responses to regulate mitochondrial ROS homeostasis and mitochondrial function, thereby enhancing epithelial defense against air pollution exposure.
Collapse
Affiliation(s)
- Ju Chang-Chien
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Jing-Long Huang
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan
| | - Hui-Ju Tsai
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
- College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Shih-Ling Wang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan
| | - Ming-Ling Kuo
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan.
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan.
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wenhua 1st Road, Kweishan, Taoyuan, 33302, Taiwan.
| | - Tsung-Chieh Yao
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Hsin Street, Kweishan, Taoyuan, 33305, Taiwan.
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan.
- Community Medicine Research Center, Chang Gung Memorial Hospital at Keelung, Keelung, Taiwan.
| |
Collapse
|
27
|
García-Poyatos C, Arora P, Calvo E, Marques IJ, Kirschke N, Galardi-Castilla M, Lembke C, Meer M, Fernández-Montes P, Ernst A, Haberthür D, Hlushchuk R, Vázquez J, Vermathen P, Enríquez JA, Mercader N. Cox7a1 controls skeletal muscle physiology and heart regeneration through complex IV dimerization. Dev Cell 2024; 59:1824-1841.e10. [PMID: 38701784 DOI: 10.1016/j.devcel.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/30/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024]
Abstract
The oxidative phosphorylation (OXPHOS) system is intricately organized, with respiratory complexes forming super-assembled quaternary structures whose assembly mechanisms and physiological roles remain under investigation. Cox7a2l, also known as Scaf1, facilitates complex III and complex IV (CIII-CIV) super-assembly, enhancing energetic efficiency in various species. We examined the role of Cox7a1, another Cox7a family member, in supercomplex assembly and muscle physiology. Zebrafish lacking Cox7a1 exhibited reduced CIV2 formation, metabolic alterations, and non-pathological muscle performance decline. Additionally, cox7a1-/- hearts displayed a pro-regenerative metabolic profile, impacting cardiac regenerative response. The distinct phenotypic effects of cox7a1-/- and cox7a2l-/- underscore the diverse metabolic and physiological consequences of impaired supercomplex formation, emphasizing the significance of Cox7a1 in muscle maturation within the OXPHOS system.
Collapse
Affiliation(s)
- Carolina García-Poyatos
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en red en Fragilidad y Envejecimiento saludable (CIBERFES), Madrid, Spain
| | - Prateek Arora
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Ines J Marques
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland
| | - Nick Kirschke
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland
| | | | - Carla Lembke
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland
| | - Marco Meer
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland
| | | | - Alexander Ernst
- Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - David Haberthür
- MicroCT research group, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Ruslan Hlushchuk
- MicroCT research group, Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Peter Vermathen
- University Institute of Diagnostic and Interventional Neuroradiology, Magnetic Resonance Methodology, University of Bern, Bern, Switzerland
| | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Centro de Investigación Biomédica en red en Fragilidad y Envejecimiento saludable (CIBERFES), Madrid, Spain.
| | - Nadia Mercader
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain; Department of Developmental Biology and Regeneration, Institute of Anatomy, University of Bern, Bern, Switzerland; Department for Biomedical Research, Cardiovascular Disease Program, University of Bern, Bern, Switzerland.
| |
Collapse
|
28
|
Mello DF, Perez L, Bergemann CM, Morton KS, Ryde IT, Meyer JN. Comprehensive characterization of mitochondrial bioenergetics at different larval stages reveals novel insights about the developmental metabolism of Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600841. [PMID: 38979262 PMCID: PMC11230424 DOI: 10.1101/2024.06.26.600841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mitochondrial bioenergetic processes are fundamental to development, stress responses, and health. Caenorhabditis elegans is widely used to study developmental biology, mitochondrial disease, and mitochondrial toxicity. Oxidative phosphorylation generally increases during development in many species, and genetic and environmental factors may alter this normal trajectory. Altered mitochondrial function during development can lead to both drastic, short-term responses including arrested development and death, and subtle consequences that may persist throughout life and into subsequent generations. Understanding normal and altered developmental mitochondrial biology in C. elegans is currently constrained by incomplete and conflicting reports on how mitochondrial bioenergetic parameters change during development in this species. We used a Seahorse XFe24 Extracellular Flux (XF) Analyzer to carry out a comprehensive analysis of mitochondrial and non-mitochondrial oxygen consumption rates (OCR) throughout larval development in C. elegans. We optimized and describe conditions for analysis of basal OCR, basal mitochondrial OCR, ATP-linked OCR, spare and maximal respiratory capacity, proton leak, and non-mitochondrial OCR. A key consideration is normalization, and we present and discuss results as normalized per individual worm, protein content, worm volume, mitochondrial DNA (mtDNA) count, nuclear DNA (ncDNA) count, and mtDNA:ncDNA ratio. Which normalization process is best depends on the question being asked, and differences in normalization explain some of the discrepancies in previously reported developmental changes in OCR in C. elegans. Broadly, when normalized to worm number, our results agree with previous reports in showing dramatic increases in OCR throughout development. However, when normalized to total protein, worm volume, or ncDNA or mtDNA count, after a significant 2-3-fold increase from L1 to L2 stages, we found small or no changes in most OCR parameters from the L2 to the L4 stage, other than a marginal increase at L3 in spare and maximal respiratory capacity. Overall, our results indicate an earlier cellular shift to oxidative metabolism than suggested in most previous literature.
Collapse
Affiliation(s)
- Danielle F. Mello
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| | - Luiza Perez
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| | - Christina M. Bergemann
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| | - Katherine S. Morton
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| | - Ian T. Ryde
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| | - Joel N. Meyer
- Nicholas School of the Environment, Duke University, Box 90328, Durham, NC, 27708-0328 United States of America
| |
Collapse
|
29
|
Hayashi Y, Okumura H, Arioka Y, Kushima I, Mori D, Lo T, Otgonbayar G, Kato H, Nawa Y, Kimura H, Aleksic B, Ozaki N. Analysis of human neuronal cells carrying ASTN2 deletion associated with psychiatric disorders. Transl Psychiatry 2024; 14:236. [PMID: 38830862 PMCID: PMC11148150 DOI: 10.1038/s41398-024-02962-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/05/2024] Open
Abstract
Recent genetic studies have found common genomic risk variants among psychiatric disorders, strongly suggesting the overlaps in their molecular and cellular mechanism. Our research group identified the variant in ASTN2 as one of the candidate risk factors across these psychiatric disorders by whole-genome copy number variation analysis. However, the alterations in the human neuronal cells resulting from ASTN2 variants identified in patients remain unknown. To address this, we used patient-derived and genome-edited iPS cells with ASTN2 deletion; cells were further differentiated into neuronal cells. A comprehensive gene expression analysis using genome-edited iPS cells with variants on both alleles revealed that the expression level of ZNF558, a gene specifically expressed in human forebrain neural progenitor cells, was greatly reduced in ASTN2-deleted neuronal cells. Furthermore, the expression of the mitophagy-related gene SPATA18, which is repressed by ZNF558, and mitophagy activity were increased in ASTN2-deleted neuronal cells. These phenotypes were also detected in neuronal cells differentiated from patient-derived iPS cells with heterozygous ASTN2 deletion. Our results suggest that ASTN2 deletion is related to the common pathogenic mechanism of psychiatric disorders by regulating mitophagy via ZNF558.
Collapse
Affiliation(s)
- Yu Hayashi
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Okumura
- Department of Hospital Pharmacy, Nagoya University Hospital, Nagoya, Japan
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuko Arioka
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan.
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Medical Genomics Center, Nagoya University Hospital, Nagoya, Japan
| | - Daisuke Mori
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Tzuyao Lo
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gantsooj Otgonbayar
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidekazu Kato
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Nawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kimura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| |
Collapse
|
30
|
Zhong J, Xu J, Chen X, Li N, Li S, Deng Z, Feng H, Ling X, Wang C, Zhou Z, Li L. Rbm46 inhibits reactive oxygen species in mouse embryonic stem cells through modulating BNIP3-mediated mitophagy. Biochem Biophys Res Commun 2024; 708:149779. [PMID: 38518724 DOI: 10.1016/j.bbrc.2024.149779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/29/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024]
Abstract
Embryonic stem cells (ESCs) exhibit a metabolic preference for glycolysis over oxidative phosphorylation to meet their substantial adenosine triphosphate (ATP) demands during self-renewal. This metabolic choice inherently maintains low mitochondrial activity and minimal reactive oxygen species (ROS) generation. Nonetheless, the intricate molecular mechanisms governing the restraint of ROS production and the mitigation of cellular damage remain incompletely elucidated. In this study, we reveal the pivotal role of RNA-binding motif protein 46 (RBM46) in ESCs, acting as a direct post transcriptional regulator of ROS levels by modulating BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (Bnip3) mRNA expression. Rbm46 knockout lead to diminished mitochondrial autophagy, culminating in elevated ROS within ESCs, disrupting the delicate balance required for healthy self-renewal. These findings provide insights into a novel mechanism governing ROS regulation in ESCs.
Collapse
Affiliation(s)
- Jinchen Zhong
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jing Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiaoyang Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Na Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Sha Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhiwen Deng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huimin Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiaohan Ling
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chenchen Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China.
| | - Zhi Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Lingsong Li
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
31
|
Jang J, Kang KW, Kim YW, Jeong S, Park J, Park J, Moon J, Jang J, Kim S, Kim S, Cho S, Lee Y, Kim HK, Han J, Ko EA, Jung SC, Kim JH, Ko JH. Cardioprotection via mitochondrial transplantation supports fatty acid metabolism in ischemia-reperfusion injured rat heart. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:209-217. [PMID: 38682169 PMCID: PMC11058541 DOI: 10.4196/kjpp.2024.28.3.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 05/01/2024]
Abstract
In addition to cellular damage, ischemia-reperfusion (IR) injury induces substantial damage to the mitochondria and endoplasmic reticulum. In this study, we sought to determine whether impaired mitochondrial function owing to IR could be restored by transplanting mitochondria into the heart under ex vivo IR states. Additionally, we aimed to provide preliminary results to inform therapeutic options for ischemic heart disease (IHD). Healthy mitochondria isolated from autologous gluteus maximus muscle were transplanted into the hearts of Sprague-Dawley rats damaged by IR using the Langendorff system, and the heart rate and oxygen consumption capacity of the mitochondria were measured to confirm whether heart function was restored. In addition, relative expression levels were measured to identify the genes related to IR injury. Mitochondrial oxygen consumption capacity was found to be lower in the IR group than in the group that underwent mitochondrial transplantation after IR injury (p < 0.05), and the control group showed a tendency toward increased oxygen consumption capacity compared with the IR group. Among the genes related to fatty acid metabolism, Cpt1b (p < 0.05) and Fads1 (p < 0.01) showed significant expression in the following order: IR group, IR + transplantation group, and control group. These results suggest that mitochondrial transplantation protects the heart from IR damage and may be feasible as a therapeutic option for IHD.
Collapse
Affiliation(s)
- Jehee Jang
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Ki-Woon Kang
- Divsion of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Young-Won Kim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Seohyun Jeong
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Jaeyoon Park
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Jihoon Park
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Jisung Moon
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Junghyun Jang
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Seohyeon Kim
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Sunghun Kim
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Sungjoo Cho
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Yurim Lee
- Department of Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, SMART Marine Therapeutics Center, Inje University, Busan 47392, Korea
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, SMART Marine Therapeutics Center, Inje University, Busan 47392, Korea
| | - Eun-A Ko
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Sung-Cherl Jung
- Department of Physiology, School of Medicine, Jeju National University, Jeju 63243, Korea
| | - Jung-Ha Kim
- Department of Family Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Jae-Hong Ko
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
32
|
Morotti M, Grimm AJ, Hope HC, Arnaud M, Desbuisson M, Rayroux N, Barras D, Masid M, Murgues B, Chap BS, Ongaro M, Rota IA, Ronet C, Minasyan A, Chiffelle J, Lacher SB, Bobisse S, Murgues C, Ghisoni E, Ouchen K, Bou Mjahed R, Benedetti F, Abdellaoui N, Turrini R, Gannon PO, Zaman K, Mathevet P, Lelievre L, Crespo I, Conrad M, Verdeil G, Kandalaft LE, Dagher J, Corria-Osorio J, Doucey MA, Ho PC, Harari A, Vannini N, Böttcher JP, Dangaj Laniti D, Coukos G. PGE 2 inhibits TIL expansion by disrupting IL-2 signalling and mitochondrial function. Nature 2024; 629:426-434. [PMID: 38658764 PMCID: PMC11078736 DOI: 10.1038/s41586-024-07352-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024]
Abstract
Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rβ-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Dinoprostone/metabolism
- Down-Regulation
- Ferroptosis
- Interleukin Receptor Common gamma Subunit/biosynthesis
- Interleukin Receptor Common gamma Subunit/deficiency
- Interleukin Receptor Common gamma Subunit/metabolism
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-2 Receptor beta Subunit/metabolism
- Lymphocytes, Tumor-Infiltrating/cytology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mitochondria/metabolism
- Oxidative Stress
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/metabolism
- Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Signal Transduction
- TOR Serine-Threonine Kinases/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Matteo Morotti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Alizee J Grimm
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Helen Carrasco Hope
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Marion Arnaud
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Mathieu Desbuisson
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Nicolas Rayroux
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - David Barras
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Maria Masid
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Baptiste Murgues
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Bovannak S Chap
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marco Ongaro
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Ioanna A Rota
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Catherine Ronet
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Aspram Minasyan
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Johanna Chiffelle
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Sebastian B Lacher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Sara Bobisse
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Clément Murgues
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Eleonora Ghisoni
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Khaoula Ouchen
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Ribal Bou Mjahed
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Fabrizio Benedetti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Naoill Abdellaoui
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Riccardo Turrini
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philippe O Gannon
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Khalil Zaman
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Patrice Mathevet
- Department of Gynaecology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Loic Lelievre
- Department of Gynaecology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Isaac Crespo
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Target and Therapeutics Centre, Helmholtz Munich, Neuherberg, Germany
| | - Gregory Verdeil
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Center of Experimental Therapeutics, Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Julien Dagher
- Unit of Translational Oncopathology, Institute of Pathology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jesus Corria-Osorio
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Marie-Agnes Doucey
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Ping-Chih Ho
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
| | - Nicola Vannini
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Denarda Dangaj Laniti
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
| | - George Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland.
- Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
| |
Collapse
|
33
|
Faura G, Studenovska H, Sekac D, Ellederova Z, Petrovski G, Eide L. The Effects of the Coating and Aging of Biodegradable Polylactic Acid Membranes on In Vitro Primary Human Retinal Pigment Epithelium Cells. Biomedicines 2024; 12:966. [PMID: 38790928 PMCID: PMC11117638 DOI: 10.3390/biomedicines12050966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Age-related macular degeneration (AMD) is the most frequent cause of blindness in developed countries. The replacement of dysfunctional human retinal pigment epithelium (hRPE) cells by the transplantation of in vitro-cultivated hRPE cells to the affected area emerges as a feasible strategy for regenerative therapy. Synthetic biomimetic membranes arise as powerful hRPE cell carriers, but as biodegradability is a requirement, it also poses a challenge due to its limited durability. hRPE cells exhibit several characteristics that putatively respond to the type of membrane carrier, and they can be used as biomarkers to evaluate and further optimize such membranes. Here, we analyze the pigmentation, transepithelial resistance, genome integrity, and maturation markers of hRPE cells plated on commercial polycarbonate (PC) versus in-house electrospun polylactide-based (PLA) membranes, both enabling separate apical/basolateral compartments. Our results show that PLA is superior to PC-based membranes for the cultivation of hRPEs, and the BEST1/RPE65 maturation markers emerge as the best biomarkers for addressing the quality of hRPE cultivated in vitro. The stability of the cultures was observed to be affected by PLA aging, which is an effect that could be partially palliated by the coating of the PLA membranes.
Collapse
Affiliation(s)
- Georgina Faura
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- CIDETEC, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastián, Spain
| | - Hana Studenovska
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, 162 00 Prague, Czech Republic;
| | - David Sekac
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
- Department of Cell Biology, Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, 277 21 Libechov, Czech Republic; (D.S.); (Z.E.)
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Oslo University Hospital and Institute for Clinical Medicine, University of Oslo, 0424 Oslo, Norway;
- Norwegian Center for Stem Cell Research, Oslo University Hospital, 0424 Oslo, Norway
- Department of Ophthalmology, University Hospital Centre, University of Split School of Medicine, 21000 Split, Croatia
- UKLO Network, University St. Kliment Ohridski, 7000 Bitola, North Macedonia
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
34
|
Lee SM, Loo CE, Prasasya RD, Bartolomei MS, Kohli RM, Zhou W. Low-input and single-cell methods for Infinium DNA methylation BeadChips. Nucleic Acids Res 2024; 52:e38. [PMID: 38407446 PMCID: PMC11040145 DOI: 10.1093/nar/gkae127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 02/27/2024] Open
Abstract
The Infinium BeadChip is the most widely used DNA methylome assay technology for population-scale epigenome profiling. However, the standard workflow requires over 200 ng of input DNA, hindering its application to small cell-number samples, such as primordial germ cells. We developed experimental and analysis workflows to extend this technology to suboptimal input DNA conditions, including ultra-low input down to single cells. DNA preamplification significantly enhanced detection rates to over 50% in five-cell samples and ∼25% in single cells. Enzymatic conversion also substantially improved data quality. Computationally, we developed a method to model the background signal's influence on the DNA methylation level readings. The modified detection P-value calculation achieved higher sensitivities for low-input datasets and was validated in over 100 000 public diverse methylome profiles. We employed the optimized workflow to query the demethylation dynamics in mouse primordial germ cells available at low cell numbers. Our data revealed nuanced chromatin states, sex disparities, and the role of DNA methylation in transposable element regulation during germ cell development. Collectively, we present comprehensive experimental and computational solutions to extend this widely used methylation assay technology to applications with limited DNA.
Collapse
Affiliation(s)
- Sol Moe Lee
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, PA 19104, USA
| | - Christian E Loo
- Graduate Group in Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rexxi D Prasasya
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marisa S Bartolomei
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rahul M Kohli
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wanding Zhou
- Center for Computational and Genomic Medicine, The Children's Hospital of Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
McShane E, Couvillion M, Ietswaart R, Prakash G, Smalec BM, Soto I, Baxter-Koenigs AR, Choquet K, Churchman LS. A kinetic dichotomy between mitochondrial and nuclear gene expression processes. Mol Cell 2024; 84:1541-1555.e11. [PMID: 38503286 PMCID: PMC11236289 DOI: 10.1016/j.molcel.2024.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/12/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024]
Abstract
Oxidative phosphorylation (OXPHOS) complexes, encoded by both mitochondrial and nuclear DNA, are essential producers of cellular ATP, but how nuclear and mitochondrial gene expression steps are coordinated to achieve balanced OXPHOS subunit biogenesis remains unresolved. Here, we present a parallel quantitative analysis of the human nuclear and mitochondrial messenger RNA (mt-mRNA) life cycles, including transcript production, processing, ribosome association, and degradation. The kinetic rates of nearly every stage of gene expression differed starkly across compartments. Compared with nuclear mRNAs, mt-mRNAs were produced 1,100-fold more, degraded 7-fold faster, and accumulated to 160-fold higher levels. Quantitative modeling and depletion of mitochondrial factors LRPPRC and FASTKD5 identified critical points of mitochondrial regulatory control, revealing that the mitonuclear expression disparities intrinsically arise from the highly polycistronic nature of human mitochondrial pre-mRNA. We propose that resolving these differences requires a 100-fold slower mitochondrial translation rate, illuminating the mitoribosome as a nexus of mitonuclear co-regulation.
Collapse
Affiliation(s)
- Erik McShane
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mary Couvillion
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Ietswaart
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gyan Prakash
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brendan M Smalec
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Iliana Soto
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Autum R Baxter-Koenigs
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Karine Choquet
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - L Stirling Churchman
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
36
|
MacDonald KM, Khan S, Lin B, Hurren R, Schimmer AD, Kislinger T, Harding SM. The proteomic landscape of genotoxic stress-induced micronuclei. Mol Cell 2024; 84:1377-1391.e6. [PMID: 38423013 DOI: 10.1016/j.molcel.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/20/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024]
Abstract
Micronuclei (MN) are induced by various genotoxic stressors and amass nuclear- and cytoplasmic-resident proteins, priming the cell for MN-driven signaling cascades. Here, we measured the proteome of micronuclear, cytoplasmic, and nuclear fractions from human cells exposed to a panel of six genotoxins, comprehensively profiling their MN protein landscape. We find that MN assemble a proteome distinct from both surrounding cytoplasm and parental nuclei, depleted of spliceosome and DNA damage repair components while enriched for a subset of the replisome. We show that the depletion of splicing machinery within transcriptionally active MN contributes to intra-MN DNA damage, a known precursor to chromothripsis. The presence of transcription machinery in MN is stress-dependent, causing a contextual induction of MN DNA damage through spliceosome deficiency. This dataset represents a unique resource detailing the global proteome of MN, guiding mechanistic studies of MN generation and MN-associated outcomes of genotoxic stress.
Collapse
Affiliation(s)
- Kate M MacDonald
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Shahbaz Khan
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Brian Lin
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Rose Hurren
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Aaron D Schimmer
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Shane M Harding
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada; Department of Radiation Oncology and Immunology, University of Toronto, Toronto, ON M5T 1P5, Canada.
| |
Collapse
|
37
|
Jahan J, Joshi S, Oca IMD, Toelle A, Lopez-Yang C, Chacon CV, Beyer AM, Garcia CA, Jarajapu YP. The role of telomerase reverse transcriptase in the mitochondrial protective functions of Angiotensin-(1-7) in diabetic CD34 + cells. Biochem Pharmacol 2024; 222:116109. [PMID: 38458330 PMCID: PMC11007670 DOI: 10.1016/j.bcp.2024.116109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/08/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Angiotensin (Ang)-(1-7) stimulates vasoprotective functions of diabetic (DB) CD34+ hematopoietic stem/progenitor cells partly by decreasing reactive oxygen species (ROS), increasing nitric oxide (NO) levels and decreasing TGFβ1 secretion. Telomerase reverse transcriptase (TERT) translocates to mitochondria and regulates ROS generation. Alternative splicing of TERT results in variants α-, β- and α-β-TERT, which may oppose functions of full-length (FL) TERT. This study tested if the protective functions of Ang-(1-7) or TGFβ1-silencing are mediated by mitoTERT and that diabetes decreases FL-TERT expression by inducing splicing. CD34+ cells were isolated from the peripheral blood mononuclear cells of nondiabetic (ND, n = 68) or DB (n = 74) subjects. NO and mitoROS levels were evaluated by flow cytometry. TERT splice variants and mitoDNA-lesions were characterized by qPCR. TRAP assay was used for telomerase activity. Decoy peptide was used to block mitochondrial translocation (mitoXTERT). TERT inhibitor or mitoXTERT prevented the effects of Ang-(1-7) on NO or mitoROS levels in DB-CD34+ cells. FL-TERT expression and telomerase activity were lower and mitoDNA-lesions were higher in DB cells compared to ND and were reversed by Ang-(1-7) or TGFβ1-silencing. The prevalence of TERT splice variants, with predominant β-TERT expression, was higher and the expression of FL-TERT was lower in DB cells (n = 25) compared to ND (n = 30). Ang-(1-7) or TGFβ1-silencing decreased TERT-splicing and increased FL-TERT. Blocking of β-splicing increased FL-TERT and protected mitoDNA in DB-cells. The findings suggest that diabetes induces TERT-splicing in CD34+ cells and that β-TERT splice variant largely contributes to the mitoDNA oxidative damage.
Collapse
Affiliation(s)
- Jesmin Jahan
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | - Shrinidh Joshi
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | - Andrew Toelle
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | | | - Andreas M Beyer
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Yagna Pr Jarajapu
- Department of Pharmaceutical Sciences, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
38
|
Gettings SM, Timbury W, Dmochowska A, Sharma R, McGonigle R, MacKenzie LE, Miquelard-Garnier G, Bourbia N. Polyethylene terephthalate (PET) micro- and nanoplastic particles affect the mitochondrial efficiency of human brain vascular pericytes without inducing oxidative stress. NANOIMPACT 2024; 34:100508. [PMID: 38663501 DOI: 10.1016/j.impact.2024.100508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024]
Abstract
The objective of this investigation was to evaluate the influence of micro- and nanoplastic particles composed of polyethylene terephthalate (PET), a significant contributor to plastic pollution, on human brain vascular pericytes. Specifically, we delved into their impact on mitochondrial functionality, oxidative stress, and the expression of genes associated with oxidative stress, ferroptosis and mitochondrial functions. Our findings demonstrate that the exposure of a monoculture of human brain vascular pericytes to PET particles in vitro at a concentration of 50 μg/ml for a duration of 3, 6 and 10 days did not elicit oxidative stress. Notably, we observed a reduction in various aspects of mitochondrial respiration, including maximal respiration, spare respiratory capacity, and ATP production in pericytes subjected to PET particles for 3 days, with a mitochondrial function recovery at 6 and 10 days. Furthermore, there were no statistically significant alterations in mitochondrial DNA copy number, or in the expression of genes linked to oxidative stress and ferroptosis, but an increase of the expression of the gene mitochondrial transcription factor A (TFAM) was noted at 3 days exposure. These outcomes suggest that, at a concentration of 50 μg/ml, PET particles do not induce oxidative stress in human brain vascular pericytes. Instead, at 3 days exposure, PET exposure impairs mitochondrial functions, but this is recovered at 6-day exposure. This seems to indicate a potential mitochondrial hormesis response (mitohormesis) is incited, involving the gene TFAM. Further investigations are warranted to explore the stages of mitohormesis and the potential consequences of plastics on the integrity of the blood-brain barrier and intercellular interactions. This research contributes to our comprehension of the potential repercussions of nanoplastic pollution on human health and underscores the imperative need for ongoing examinations into the exposure to plastic particles.
Collapse
Affiliation(s)
- Sean M Gettings
- UK Health Security Agency, Radiation Effects Department, Radiation Protection Science Division, Harwell Science Campus, Didcot, Oxfordshire OX11 0RQ, UK
| | - William Timbury
- UK Health Security Agency, Radiation Effects Department, Radiation Protection Science Division, Harwell Science Campus, Didcot, Oxfordshire OX11 0RQ, UK
| | - Anna Dmochowska
- Laboratoire PIMM, CNRS, Arts et Métiers Institute of Technology, Cnam, HESAM Universite, 75013 Paris, France
| | - Riddhi Sharma
- UK Health Security Agency, Radiation Effects Department, Radiation Protection Science Division, Harwell Science Campus, Didcot, Oxfordshire OX11 0RQ, UK
| | - Rebecca McGonigle
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1RD, UK
| | - Lewis E MacKenzie
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1RD, UK
| | - Guillaume Miquelard-Garnier
- Laboratoire PIMM, CNRS, Arts et Métiers Institute of Technology, Cnam, HESAM Universite, 75013 Paris, France
| | - Nora Bourbia
- UK Health Security Agency, Radiation Effects Department, Radiation Protection Science Division, Harwell Science Campus, Didcot, Oxfordshire OX11 0RQ, UK.
| |
Collapse
|
39
|
Rathor L, Curry S, Park Y, McElroy T, Robles B, Sheng Y, Chen WW, Min K, Xiao R, Lee MH, Han SM. Mitochondrial stress in GABAergic neurons non-cell autonomously regulates organismal health and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585932. [PMID: 38585797 PMCID: PMC10996468 DOI: 10.1101/2024.03.20.585932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Mitochondrial stress within the nervous system can trigger non-cell autonomous responses in peripheral tissues. However, the specific neurons involved and their impact on organismal aging and health have remained incompletely understood. Here, we demonstrate that mitochondrial stress in γ-aminobutyric acid-producing (GABAergic) neurons in Caenorhabditis elegans ( C. elegans ) is sufficient to significantly alter organismal lifespan, stress tolerance, and reproductive capabilities. This mitochondrial stress also leads to significant changes in mitochondrial mass, energy production, and levels of reactive oxygen species (ROS). DAF-16/FoxO activity is enhanced by GABAergic neuronal mitochondrial stress and mediates the induction of these non-cell-autonomous effects. Moreover, our findings indicate that GABA signaling operates within the same pathway as mitochondrial stress in GABAergic neurons, resulting in non-cell-autonomous alterations in organismal stress tolerance and longevity. In summary, these data suggest the crucial role of GABAergic neurons in detecting mitochondrial stress and orchestrating non-cell-autonomous changes throughout the organism.
Collapse
|
40
|
Kodavati M, Wang H, Guo W, Mitra J, Hegde PM, Provasek V, Rao VHM, Vedula I, Zhang A, Mitra S, Tomkinson AE, Hamilton DJ, Van Den Bosch L, Hegde ML. FUS unveiled in mitochondrial DNA repair and targeted ligase-1 expression rescues repair-defects in FUS-linked motor neuron disease. Nat Commun 2024; 15:2156. [PMID: 38461154 PMCID: PMC10925063 DOI: 10.1038/s41467-024-45978-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 02/08/2024] [Indexed: 03/11/2024] Open
Abstract
This study establishes the physiological role of Fused in Sarcoma (FUS) in mitochondrial DNA (mtDNA) repair and highlights its implications to the pathogenesis of FUS-associated neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). Endogenous FUS interacts with and recruits mtDNA Ligase IIIα (mtLig3) to DNA damage sites within mitochondria, a relationship essential for maintaining mtDNA repair and integrity in healthy cells. Using ALS patient-derived FUS mutant cell lines, a transgenic mouse model, and human autopsy samples, we discovered that compromised FUS functionality hinders mtLig3's repair role, resulting in increased mtDNA damage and mutations. These alterations cause various manifestations of mitochondrial dysfunction, particularly under stress conditions relevant to disease pathology. Importantly, rectifying FUS mutations in patient-derived induced pluripotent cells (iPSCs) preserves mtDNA integrity. Similarly, targeted introduction of human DNA Ligase 1 restores repair mechanisms and mitochondrial activity in FUS mutant cells, suggesting a potential therapeutic approach. Our findings unveil FUS's critical role in mitochondrial health and mtDNA repair, offering valuable insights into the mechanisms underlying mitochondrial dysfunction in FUS-associated motor neuron disease.
Collapse
Affiliation(s)
- Manohar Kodavati
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Haibo Wang
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Wenting Guo
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- INSERM, UMR-S1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, Université de Strasbourg, CRBS, Strasbourg, France
| | - Joy Mitra
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Pavana M Hegde
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Vincent Provasek
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
- College of Medicine, Texas A&M University, College Station, TX, USA
| | - Vikas H Maloji Rao
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Indira Vedula
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Aijun Zhang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine affiliate, Houston, TX, USA
| | - Sankar Mitra
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA
| | - Alan E Tomkinson
- Departments of Internal Medicine, and Molecular Genetics and Microbiology and University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Dale J Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
- Department of Medicine, Houston Methodist, Weill Cornell Medicine affiliate, Houston, TX, USA
| | - Ludo Van Den Bosch
- KU Leuven-Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Muralidhar L Hegde
- Division of DNA Repair Research within the Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Neuroscience, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
41
|
Li H, Gao W, Wang H, Zhang H, Huang L, Yuan T, Zheng W, Wu Q, Liu J, Xu W, Wang W, Yang L, Zhu Y. Evidence from an Avian Embryo Model that Zinc-Inducible MT4 Expression Protects Mitochondrial Function Against Oxidative Stress. J Nutr 2024; 154:896-907. [PMID: 38301957 DOI: 10.1016/j.tjnut.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Metallothioneins (MTs) have a strong affinity for zinc (Zn) and remain at a sufficiently high level in mitochondria. As the avian embryo is highly susceptible to oxidative damage and relatively easy to manipulate in a naturally closed chamber, it is an ideal model of the effects of oxidative stress on mitochondrial function. However, the protective roles and molecular mechanisms of Zn-inducible protein expression on mitochondrial function in response to various stressors are poorly understood. OBJECTIVES The study aimed to investigate the mechanisms by which Zn-induced MT4 expression protects mitochondrial function and energy metabolism subjected to oxidative stress using the avian embryo and embryonic primary hepatocyte models. METHODS First, we investigated whether MT4 expression alters mitochondrial function. Then, we examined the effects of Zn-induced MT4 overexpression and MT4 silencing on embryonic primary hepatocytes from breeder hens fed a normal Zn diet subjected to a tert-butyl hydroperoxide (BHP) oxidative stress challenge during incubation. In vivo, the avian embryos from hens fed the Zn-deficient and Zn-adequate diets were used to determine the protective roles of Zn-induced MT4 expression on the function of mitochondria exposed to oxidative stress induced by in ovo BHP injection. RESULTS An in vitro study revealed that Zn-induced MT4 expression reduced reactive oxygen species accumulation in primary hepatocytes. MT4 silencing exacerbated BHP-mediated mitochondrial dysfunction whereas Zn-inducible MT4 overexpression mitigated it. Another in vivo study disclosed that maternal Zn-induced MT4 expression protected mitochondrial function in chick embryo hepatocytes against oxidative stress by inhibiting the peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α)/peroxisome proliferators-activated receptor-γ (PPAR-γ) pathway. CONCLUSION This study underscores the potential protective roles of Zn-induced MT4 expression via the downregulation of the PGC-1α/PPAR-γ pathway on mitochondrial function stimulated by the stress challenge in the primary hepatocytes in an avian embryo model. Our findings suggested that Zn-induced MT4 expression could provide a new therapeutic target and preventive strategy for repairing mitochondrial dysfunction in disease.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Wei Gao
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Heng Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Huaqi Zhang
- College of Agriculture, Tongren Polytechnic University, Tongren, People's Republic of China
| | - Liang Huang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Tong Yuan
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Wenxuan Zheng
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Qilin Wu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Ju Liu
- Department of Poultry Breeding, Enping Long Industrial Co. Ltd., Enping, People's Republic of China
| | - Weihan Xu
- Department of Poultry Breeding, Zhengzhi Poultry Industry Co. Ltd., Shantou, People's Republic of China
| | - Wence Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China
| | - Lin Yang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.
| | - Yongwen Zhu
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou, People's Republic of China.
| |
Collapse
|
42
|
Isaac RS, Tullius TW, Hansen KG, Dubocanin D, Couvillion M, Stergachis AB, Churchman LS. Single-nucleoid architecture reveals heterogeneous packaging of mitochondrial DNA. Nat Struct Mol Biol 2024; 31:568-577. [PMID: 38347148 PMCID: PMC11370055 DOI: 10.1038/s41594-024-01225-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/09/2024] [Indexed: 03/03/2024]
Abstract
Cellular metabolism relies on the regulation and maintenance of mitochondrial DNA (mtDNA). Hundreds to thousands of copies of mtDNA exist in each cell, yet because mitochondria lack histones or other machinery important for nuclear genome compaction, it remains unresolved how mtDNA is packaged into individual nucleoids. In this study, we used long-read single-molecule accessibility mapping to measure the compaction of individual full-length mtDNA molecules at near single-nucleotide resolution. We found that, unlike the nuclear genome, human mtDNA largely undergoes all-or-none global compaction, with most nucleoids existing in an inaccessible, inactive state. Highly accessible mitochondrial nucleoids are co-occupied by transcription and replication components and selectively form a triple-stranded displacement loop structure. In addition, we showed that the primary nucleoid-associated protein TFAM directly modulates the fraction of inaccessible nucleoids both in vivo and in vitro, acting consistently with a nucleation-and-spreading mechanism to coat and compact mitochondrial nucleoids. Together, these findings reveal the primary architecture of mtDNA packaging and regulation in human cells.
Collapse
Affiliation(s)
- R Stefan Isaac
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Thomas W Tullius
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Katja G Hansen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Danilo Dubocanin
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mary Couvillion
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| | - L Stirling Churchman
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
43
|
Veilleux V, Pichaud N, Boudreau LH, Robichaud GA. Mitochondria Transfer by Platelet-Derived Microparticles Regulates Breast Cancer Bioenergetic States and Malignant Features. Mol Cancer Res 2024; 22:268-281. [PMID: 38085263 DOI: 10.1158/1541-7786.mcr-23-0329] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/25/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024]
Abstract
An increasing number of studies show that platelets as well as platelet-derived microparticles (PMP) play significant roles in cancer malignancy and disease progression. Particularly, PMPs have the capacity to interact and internalize within target cells resulting in the transfer of their bioactive cargo, which can modulate the signaling and activation processes of recipient cells. We recently identified a new subpopulation of these vesicles (termed mitoMPs), which contain functional mitochondria. Given the predominant role of mitochondria in cancer cell metabolism and disease progression, we set out to investigate the impact of mitoMPs on breast cancer metabolic reprograming and phenotypic processes leading to malignancy. Interestingly, we observed that recipient cell permeability to PMP internalization varied among the breast cancer cell types evaluated in our study. Specifically, cells permissive to mitoMPs acquire mitochondrial-dependent functions, which stimulate increased cellular oxygen consumption rates and intracellular ATP levels. In addition, cancer cells co-incubated with PMPs display enhanced malignant features in terms of migration and invasion. Most importantly, the cancer aggressive processes and notable metabolic plasticity induced by PMPs were highly dependent on the functional status of the mitoMP-packaged mitochondria. These findings characterize a new mechanism by which breast cancer cells acquire foreign mitochondria resulting in the gain of metabolic processes and malignant features. A better understanding of these mechanisms may provide therapeutic opportunities through PMP blockade to deprive cancer cells from resources vital in disease progression. IMPLICATIONS We show that the transfer of foreign mitochondria by microparticles modulates recipient cancer cell metabolic plasticity, leading to greater malignant processes.
Collapse
Affiliation(s)
- Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| |
Collapse
|
44
|
Nakajima KI, Inagaki T, Espera JM, Izumiya Y. Kaposi's sarcoma-associated herpesvirus (KSHV) LANA prevents KSHV episomes from degradation. J Virol 2024; 98:e0126823. [PMID: 38240588 PMCID: PMC10878079 DOI: 10.1128/jvi.01268-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/14/2023] [Indexed: 02/21/2024] Open
Abstract
Protein knockdown with an inducible degradation system is a powerful tool for studying proteins of interest in living cells. Here, we adopted the auxin-inducible degron (AID) approach to detail Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA) function in latency maintenance and inducible viral lytic gene expression. We fused the mini-auxin-inducible degron (mAID) tag at the LANA N-terminus with KSHV bacterial artificial chromosome 16 recombination, and iSLK cells were stably infected with the recombinant KSHV encoding mAID-LANA. Incubation with 5-phenyl-indole-3-acetic acid, a derivative of natural auxin, rapidly degraded LANA within 1.5 h. In contrast to our hypothesis, depletion of LANA alone did not trigger lytic reactivation but rather decreased inducible lytic gene expression when we stimulated reactivation with a combination of ORF50 protein expression and sodium butyrate. Decreased overall lytic gene induction seemed to be associated with a rapid loss of KSHV genomes in the absence of LANA. The rapid loss of viral genomic DNA was blocked by a lysosomal inhibitor, chloroquine. Furthermore, siRNA-mediated knockdown of cellular innate immune proteins, cyclic AMP-GMP synthase (cGAS) and simulator of interferon genes (STING), and other autophagy-related genes rescued the degradation of viral genomic DNA upon LANA depletion. Reduction of the viral genome was not observed in 293FT cells that lack the expression of cGAS. These results suggest that LANA actively prevents viral genomic DNA from sensing by cGAS-STING signaling axis, adding novel insights into the role of LANA in latent genome maintenance.IMPORTANCESensing of pathogens' components is a fundamental cellular immune response. Pathogens have therefore evolved strategies to evade such cellular immune responses. KSHV LANA is a multifunctional protein and plays an essential role in maintaining the latent infection by tethering viral genomic DNA to the host chromosome. We adopted the inducible protein knockdown approach and found that depletion of LANA induced rapid degradation of viral genomic DNA, which is mediated by innate immune DNA sensors and autophagy pathway. These observations suggest that LANA may play a role in hiding KSHV episome from innate immune DNA sensors. Our study thus provides new insights into the role of LANA in latency maintenance.
Collapse
Affiliation(s)
- Ken-ichi Nakajima
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Tomoki Inagaki
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Jonna Magdallene Espera
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, University of California Davis, Sacramento, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California, USA
| |
Collapse
|
45
|
Llavanera M, Mateo-Otero Y, Viñolas-Vergés E, Bonet S, Yeste M. Sperm function, mitochondrial activity and in vivo fertility are associated to their mitochondrial DNA content in pigs. J Anim Sci Biotechnol 2024; 15:10. [PMID: 38297401 PMCID: PMC10832242 DOI: 10.1186/s40104-023-00988-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Despite their low abundance in sperm, mitochondria have diverse functions in this cell type, including energy production, signalling and calcium regulation. In humans, sperm mitochondrial DNA content (mtDNAc) has been reported to be negatively linked to sperm function and fertility. Yet, the association between mtDNAc and sperm function in livestock remains unexplored. For this reason, this study aimed to shed some light on the link between mtDNAc and sperm function and fertilising potential in pigs. A qPCR method for mtDNAc quantification was optimised for pig sperm, and the association of this parameter with sperm motility, kinematics, mitochondrial activity, and fertility was subsequently interrogated. RESULTS First, the qPCR method was found to be sensitive and efficient for mtDNAc quantification in pig sperm. By using this technique, mtDNAc was observed to be associated to sperm motility, mitochondrial activity and in vivo, but not in vitro, fertility outcomes. Specifically, sperm with low mtDNAc were seen to exhibit greater motility but decreased mitochondrial activity and intracellular reactive oxygen species. Interestingly, samples with lower mtDNAc showed higher conception and farrowing rates, but similar in vitro fertilisation rates and embryo development, when compared to those with greater mtDNAc. CONCLUSIONS These findings enrich our comprehension of the association of mtDNAc with sperm biology, and lay the foundation for future research into employing this parameter as a molecular predictor for sperm function and fertility in livestock.
Collapse
Affiliation(s)
- Marc Llavanera
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, ES-17003, Spain.
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, ES-17003, Spain.
| | - Yentel Mateo-Otero
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, ES-17003, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, ES-17003, Spain
| | - Estel Viñolas-Vergés
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, ES-17003, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, ES-17003, Spain
| | - Sergi Bonet
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, ES-17003, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, ES-17003, Spain
| | - Marc Yeste
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, ES-17003, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, ES-17003, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, ES-08010, Spain
| |
Collapse
|
46
|
Pantaleão LC, Loche E, Fernandez-Twinn DS, Dearden L, Córdova-Casanova A, Osmond C, Salonen MK, Kajantie E, Niu Y, de Almeida-Faria J, Thackray BD, Mikkola TM, Giussani DA, Murray AJ, Bushell M, Eriksson JG, Ozanne SE. Programming of cardiac metabolism by miR-15b-5p, a miRNA released in cardiac extracellular vesicles following ischemia-reperfusion injury. Mol Metab 2024; 80:101875. [PMID: 38218535 PMCID: PMC10832484 DOI: 10.1016/j.molmet.2024.101875] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVE We investigated the potential involvement of miRNAs in the developmental programming of cardiovascular diseases (CVD) by maternal obesity. METHODS Serum miRNAs were measured in individuals from the Helsinki Birth Cohort (with known maternal body mass index), and a mouse model was used to determine causative effects of maternal obesity during pregnancy and ischemia-reperfusion on offspring cardiac miRNA expression and release. RESULTS miR-15b-5p levels were increased in the sera of males born to mothers with higher BMI and in the hearts of adult mice born to obese dams. In an ex-vivo model of perfused mouse hearts, we demonstrated that cardiac tissue releases miR-15b-5p, and that some of the released miR-15b-5p was contained within small extracellular vesicles (EVs). We also demonstrated that release was higher from hearts exposed to maternal obesity following ischaemia/reperfusion. Over-expression of miR-15b-5p in vitro led to loss of outer mitochondrial membrane stability and to repressed fatty acid oxidation in cardiomyocytes. CONCLUSIONS These findings suggest that miR-15-b could play a mechanistic role in the dysregulation of cardiac metabolism following exposure to an in utero obesogenic environment and that its release in cardiac EVs following ischaemic damage may be a novel factor contributing to inter-organ communication between the programmed heart and peripheral tissues.
Collapse
Affiliation(s)
- Lucas C Pantaleão
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Elena Loche
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Laura Dearden
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Adriana Córdova-Casanova
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK
| | - Minna K Salonen
- Finnish Institute for Health and Welfare, Public Health Unit, Finland
| | - Eero Kajantie
- Finnish Institute for Health and Welfare, Public Health Unit, Finland; Clinical Medicine Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Youguo Niu
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Juliana de Almeida-Faria
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Benjamin D Thackray
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tuija M Mikkola
- Finnish Institute for Health and Welfare, Public Health Unit, Finland; Folkhalsan Research Center, Helsinki, Finland; Faculty of Medicine, University of Helsinki, Finland
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Johan G Eriksson
- Folkhalsan Research Center, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Finland; Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Susan E Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
47
|
Sarabhai T, Kahl S, Gancheva S, Mastrototaro L, Dewidar B, Pesta D, Ratter-Rieck JM, Bobrov P, Jeruschke K, Esposito I, Schlensak M, Roden M. Loss of mitochondrial adaptation associates with deterioration of mitochondrial turnover and structure in metabolic dysfunction-associated steatotic liver disease. Metabolism 2024; 151:155762. [PMID: 38122893 DOI: 10.1016/j.metabol.2023.155762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/29/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Obesity and type 2 diabetes frequently have metabolic dysfunction-associated steatotic liver disease (MASLD) including steatohepatitis (MASH). In obesity, the liver may adapt its oxidative capacity, but the role of mitochondrial turnover in MASLD remains uncertain. METHODS This cross-sectional study compared individuals with class III obesity (n = 8/group) without (control, OBE CON; NAFLD activity score: 0.4 ± 0.1) or with steatosis (OBE MASL, 2.3 ± 0.4), or MASH (OBE MASH, 5.3 ± 0.3, p < 0.05 vs. other groups). Hepatic mitochondrial ultrastructure was assessed by transmission electron microscopy, mitochondrial respiration by high-resolution respirometry, biomarkers of mitochondrial quality control and endoplasmic reticulum (ER) stress by Western Blot. RESULTS Mitochondrial oxidative capacity was 31 % higher in OBE MASL, but 25 % lower in OBE MASH (p < 0.05 vs. OBE CON). OBE MASH showed ~1.5fold lower mitochondrial number, but ~1.2-1.5fold higher diameter and area (p < 0.001 vs. other groups). Biomarkers of autophagy (p62), mitophagy (PINK1, PARKIN), fission (DRP-1, FIS1) and fusion (MFN1/2, OPA1) were reduced in OBE MASH (p < 0.05 vs. OBE CON). OBE MASL showed lower p62, p-PARKIN/PARKIN, and p-DRP-1 (p < 0.05 vs. OBE CON). OBE MASL and MASH showed higher ER stress markers (PERK, ATF4, p-eIF2α-S51/eIF2α; p < 0.05 vs. OBE CON). Mitochondrial diameter associated inversely with fusion/fission biomarkers and with oxidative capacity, but positively with H2O2. CONCLUSION Humans with hepatic steatosis already exhibit impaired mitochondrial turnover, despite upregulated oxidative capacity, and evidence for ER stress. In MASH, oxidative stress likely mediates progressive decline of mitochondrial turnover, ultrastructure and respiration indicating that mitochondrial quality control is key for energy metabolism and may have potential for targeting MASH. ClinGovTrial:NCT01477957.
Collapse
Affiliation(s)
- Theresia Sarabhai
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sabine Kahl
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Sofiya Gancheva
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Bedair Dewidar
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Dominik Pesta
- Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany; Centre for Endocrinology, Diabetes and Preventive Medicine, University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - Jacqueline M Ratter-Rieck
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Pavel Bobrov
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany; Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany
| | - Kay Jeruschke
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany
| | - Irene Esposito
- Institute of Pathology, University Hospital and Heinrich-Heine-University, Düsseldorf, Germany
| | - Matthias Schlensak
- Department of Obesity and Reflux Center, Neuwerk Hospital Mönchengladbach, Germany
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Germany; German Center for Diabetes Research, Partner Düsseldorf, Neuherberg, Germany.
| |
Collapse
|
48
|
Cereceda L, Cardenas JC, Khoury M, Silva-Pavez E, Hidalgo Y. Impact of platelet-derived mitochondria transfer in the metabolic profiling and progression of metastatic MDA-MB-231 human triple-negative breast cancer cells. Front Cell Dev Biol 2024; 11:1324158. [PMID: 38283990 PMCID: PMC10811077 DOI: 10.3389/fcell.2023.1324158] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction: An active role of platelets in the progression of triple-negative breast cancer (TNBC) cells has been described. Even the role of platelet-derived extracellular vesicles on the migration of MDA-MB-231 cells has been reported. Interestingly, upon activation, platelets release functional mitochondria into the extracellular environment. However, the impact of these platelet-derived mitochondria on the metabolic properties of MDA-MB-231 cells remains unclear. Methods: MDA-MB-231 and MDA-MB-231-Rho-0 cells were co-cultured with platelets, which were isolated from donor blood. Mitochondrial transfer was assessed through confocal microscopy and flow cytometry, while metabolic analyses were conducted using a Seahorse XF HS Mini Analyzer. The mito-chondrial DNA (mtDNA) copy number was determined via quantitative PCR (qPCR) following platelet co-culture. Finally, cell proliferation and colony formation assay were performed using crystal violet staining. Results and Discussion: We have shown that platelet-derived mitochondria are internalized by MDA-MB-231 cells in co-culture with platelets, increasing ATP production, oxygen (O2) consumption rate (OCR), cell proliferation, and metabolic adaptability. Additionally, we observed that MDA-MB-231 cells depleted from mtDNA restore cell proliferation in uridine/pyruvate-free cell culture medium and mitochondrial O2 consumption after co-culture with platelets, indicating a reconstitution of mtDNA facilitated by platelet-derived mitochondria. In conclusion, our study provides new insights into the role of platelet-derived mitochondria in the metabolic adaptability and progression of metastatic MDA-MB-231 TNBC cells.
Collapse
Affiliation(s)
- Lucas Cereceda
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - J. Cesar Cardenas
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Maroun Khoury
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
- Cells for Cells and Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Eduardo Silva-Pavez
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Bellavista, Santiago, Chile
| | - Yessia Hidalgo
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
49
|
Motta G, Thangaraj SV, Padmanabhan V. Developmental Programming: Impact of Prenatal Exposure to Bisphenol A on Senescence and Circadian Mediators in the Liver of Sheep. TOXICS 2023; 12:15. [PMID: 38250971 PMCID: PMC10818936 DOI: 10.3390/toxics12010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024]
Abstract
Prenatal exposure to endocrine disruptors such as bisphenol A (BPA) plays a critical role in the developmental programming of liver dysfunction that is characteristic of nonalcoholic fatty liver disease (NAFLD). Circadian and aging processes have been implicated in the pathogenesis of NAFLD. We hypothesized that the prenatal BPA-induced fatty-liver phenotype of female sheep is associated with premature hepatic senescence and disruption in circadian clock genes. The expression of circadian rhythm and aging-associated genes, along with other markers of senescence such as telomere length, mitochondrial DNA copy number, and lipofuscin accumulation, were evaluated in the liver tissue of control and prenatal BPA groups. Prenatal BPA exposure significantly elevated the expression of aging-associated genes GLB1 and CISD2 and induced large magnitude differences in the expression of other aging genes-APOE, HGF, KLOTHO, and the clock genes PER2 and CLOCK-in the liver; the other senescence markers remained unaffected. Prenatal BPA-programmed aging-related transcriptional changes in the liver may contribute to pathological changes in liver function, elucidating the involvement of aging genes in the pathogenesis of liver steatosis.
Collapse
Affiliation(s)
| | | | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48105, USA; (G.M.); (S.V.T.)
| |
Collapse
|
50
|
Sikder MM, Uyama T, Sasaki S, Kawai K, Araki N, Ueda N. PLAAT1 expression triggers fragmentation of mitochondria in an enzyme activity-dependent manner. J Biochem 2023; 175:101-113. [PMID: 37818970 DOI: 10.1093/jb/mvad079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
The phospholipase A and acyltransferase (PLAAT) family is a protein family consisting of five members (PLAAT1-5), which acts as phospholipid-metabolizing enzymes with phospholipase A1/A2 and N-acyltransferase activities. Since we previously reported that the overexpression of PLAAT3 in mammalian cells causes the specific disappearance of peroxisomes, in the present study we examined a possible effect of PLAAT1 on organelles. We prepared HEK293 cells expressing mouse PLAAT1 in a doxycycline-dependent manner and found that the overexpression of PLAAT1 resulted in the transformation of mitochondria from the original long rod shape to a round shape, as well as their fragmentation. In contrast, the overexpression of a catalytically inactive point mutant of PLAAT1 did not generate any morphological change in mitochondria, suggesting the involvement of catalytic activity. PLAAT1 expression also caused the reduction of peroxisomes, while the levels of the marker proteins for ER, Golgi apparatus and lysosomes were almost unchanged. In PLAAT1-expressing cells, the level of dynamin-related protein 1 responsible for mitochondrial fission was increased, whereas those of optic atrophy 1 and mitofusin 2, both of which are responsible for mitochondrial fusion, were reduced. These results suggest a novel role of PLAAT1 in the regulation of mitochondrial biogenesis.
Collapse
Affiliation(s)
- Mohammad Mamun Sikder
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Toru Uyama
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Sumire Sasaki
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Katsuhisa Kawai
- Department of Histology and Cell Biology, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Nobukazu Araki
- Department of Histology and Cell Biology, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| | - Natsuo Ueda
- Department of Biochemistry, Kagawa University School of Medicine, 1750-1 Ikenobe, Miki, Kagawa 761-0793, Japan
| |
Collapse
|