1
|
Kim HS, Sanchez ML, Silva J, Schubert HL, Dennis R, Hill CP, Christian JL. Mutations that prevent phosphorylation of the BMP4 prodomain impair proteolytic maturation of homodimers leading to lethality in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.08.617306. [PMID: 39416136 PMCID: PMC11482978 DOI: 10.1101/2024.10.08.617306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Bone morphogenetic protein4 (BMP4) plays numerous roles during embryogenesis and can signal either alone as a homodimer, or together with BMP7 as a more active heterodimer. BMPs are generated as inactive precursor proteins that dimerize and are cleaved to generate the bioactive ligand and inactive prodomain fragments. In humans, heterozygous mutations within the prodomain of BMP4 are associated with birth defects. We studied the effect of two of these mutations (p.S91C and p.E93G), which disrupt a conserved FAM20C phosphorylation motif, on ligand activity. We compared the activity of ligands generated from BMP4, BMP4S91C or BMP4E93G in Xenopus embryos and found that these mutations reduce the activity of BMP4 homodimers but not BMP4/7 heterodimers. We generated Bmp4S91C and Bmp4E93G knock-in mice and found that Bmp4S91C/S91C mice die by E11.5 and display reduced BMP activity in multiple tissues including the heart. Most Bmp4E93G/E93G mice die before weaning and Bmp4-/E93G mutants die prenatally with reduced or absent eyes, heart and ventral body wall closure defects. Mouse embryonic fibroblasts (MEFs) isolated from Bmp4S91C and Bmp4E93G embryos show accumulation of BMP4 precursor protein, reduced levels of cleaved BMP ligand and reduced BMP activity relative to MEFs from wild type littermates. Because Bmp7 is not expressed in MEFs, the accumulation of unprocessed BMP4 precursor protein in mice carrying these mutations most likely reflects an inability to cleave BMP4 homodimers, leading to reduced levels of ligand and BMP activity in vivo. Our results suggest that phosphorylation of the BMP4 prodomain is required for proteolytic activation of BMP4 homodimers, but not heterodimers.
Collapse
|
2
|
Novak J, Nahacka Z, Oliveira GL, Brisudova P, Dubisova M, Dvorakova S, Miklovicova S, Dalecka M, Puttrich V, Grycova L, Magalhaes-Novais S, Correia CM, Levoux J, Stepanek L, Prochazka J, Svec D, Reguera DP, Lopez-Domenech G, Zobalova R, Sedlacek R, Terp MG, Gammage PA, Lansky Z, Kittler J, Oliveira PJ, Ditzel HJ, Berridge MV, Rodriguez AM, Boukalova S, Rohlena J, Neuzil J. The adaptor protein Miro1 modulates horizontal transfer of mitochondria in mouse melanoma models. Cell Rep 2025; 44:115154. [PMID: 39792553 DOI: 10.1016/j.celrep.2024.115154] [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: 10/31/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Recent research has shown that mtDNA-deficient cancer cells (ρ0 cells) acquire mitochondria from tumor stromal cells to restore respiration, facilitating tumor formation. We investigated the role of Miro1, an adaptor protein involved in movement of mitochondria along microtubules, in this phenomenon. Inducible Miro1 knockout (Miro1KO) mice markedly delayed tumor formation after grafting ρ0 cancer cells. Miro1KO mice with fluorescently labeled mitochondria revealed that this delay was due to hindered mitochondrial transfer from the tumor stromal cells to grafted B16 ρ0 cells, which impeded recovery of mitochondrial respiration and tumor growth. Miro1KO led to the perinuclear accumulation of mitochondria and impaired mobility of the mitochondrial network. In vitro experiments revealed decreased association of mitochondria with microtubules, compromising mitochondrial transfer via tunneling nanotubes (TNTs) in mesenchymal stromal cells. Here we show the role of Miro1 in horizontal mitochondrial transfer in mouse melanoma models in vivo and its involvement with TNTs.
Collapse
Affiliation(s)
- Jaromir Novak
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic.
| | - Gabriela L Oliveira
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; NC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197 Cantanhede, Portugal; CIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; Institute for Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3060-197 Cantanhede, Portugal
| | - Petra Brisudova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Maria Dubisova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Sarka Dvorakova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Sona Miklovicova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Marketa Dalecka
- Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Verena Puttrich
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Lenka Grycova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Silvia Magalhaes-Novais
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Czech Center for Phenogenomic, Institute of Molecular Genetics, Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | | | - Jennifer Levoux
- Sorbonne University, Institute of Biology Paris-Seine, 75005 Paris, France
| | - Ludek Stepanek
- Czech Center for Phenogenomic, Institute of Molecular Genetics, Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Jan Prochazka
- Czech Center for Phenogenomic, Institute of Molecular Genetics, Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - David Svec
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - David Pajuelo Reguera
- Czech Center for Phenogenomic, Institute of Molecular Genetics, Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Guillermo Lopez-Domenech
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Radek Sedlacek
- Czech Center for Phenogenomic, Institute of Molecular Genetics, Czech Academy of Sciences, 252 50 Vestec, Czech Republic
| | - Mikkel G Terp
- Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Payam A Gammage
- Cancer Research UK Scotland Institute, Glasgow G61 1BD, UK; School of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Josef Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Paulo J Oliveira
- NC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, 3060-197 Cantanhede, Portugal; CIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal
| | - Henrik J Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5000 Odense, Denmark
| | | | - Anne-Marie Rodriguez
- Sorbonne University, Institute of Biology Paris-Seine, 75005 Paris, France; University Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France
| | - Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50 Prague-West, Czech Republic; Faculty of Science, Charles University, 128 00 Prague, Czech Republic; School of Pharmacy and Medical Science, Griffith University, Southport, QLD 4222, Australia; 1(st) Faculty of Medicine, Charles University, 121 08 Prague, Czech Republic.
| |
Collapse
|
3
|
Kolapalli SP, Beese CJ, Reid SE, Brynjólfsdóttir SH, Jørgensen MH, Jain A, Cuenco J, Lewinska M, Abdul-Al A, López AR, Jäättelä M, Sakamoto K, Andersen JB, Maeda K, Rusten TE, Lund AH, Frankel LB. Pellino 3 E3 ligase promotes starvation-induced autophagy to prevent hepatic steatosis. SCIENCE ADVANCES 2025; 11:eadr2450. [PMID: 39823344 PMCID: PMC11740972 DOI: 10.1126/sciadv.adr2450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Nutrient deprivation is a major trigger of autophagy, a conserved quality control and recycling process essential for cellular and tissue homeostasis. In a high-content image-based screen of the human ubiquitome, we here identify the E3 ligase Pellino 3 (PELI3) as a crucial regulator of starvation-induced autophagy. Mechanistically, PELI3 localizes to autophagic membranes, where it interacts with the ATG8 proteins through an LC3-interacting region (LIR). This facilitates PELI3-mediated ubiquitination of ULK1, driving ULK1's subsequent proteasomal degradation. PELI3 depletion leads to an aberrant accumulation and mislocalization of ULK1 and disrupts the early steps of autophagosome formation. Genetic deletion of Peli3 in mice impairs fasting-induced autophagy in the liver and enhances starvation-induced hepatic steatosis by reducing autophagy-mediated clearance of lipid droplets. Notably, PELI3 expression is decreased in the livers of patients with metabolic dysfunction-associated steatotic liver disease (MASLD), suggesting its role in hepatic steatosis development in humans. The findings suggest that PELI3-mediated control of autophagy plays a protective role in liver health.
Collapse
Affiliation(s)
- Srinivasa P. Kolapalli
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Carsten J. Beese
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Steven E. Reid
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | | | - Maria H. Jørgensen
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Ashish Jain
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Joyceline Cuenco
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Monika Lewinska
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Gubra, DK-2970 Hørsholm, Denmark
| | - Ahmad Abdul-Al
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Aida R. López
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Tor E. Rusten
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anders H. Lund
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Lisa B. Frankel
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Rao Y, Qin C, Savas AC, Liu Q, Feng S, Hou G, Xie T, Feng P. Pyrimidine synthesis enzyme CTP synthetase 1 suppresses antiviral interferon induction by deamidating IRF3. Immunity 2025; 58:74-89.e6. [PMID: 39719712 PMCID: PMC11735333 DOI: 10.1016/j.immuni.2024.11.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/14/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024]
Abstract
Metabolism is typically contextualized in conjunction with proliferation and growth. The roles of metabolic enzymes beyond metabolism-such as in innate immune responses-are underexplored. Using a focused short hairpin RNA (shRNA)-mediated screen, we identified CTP synthetase 1 (CTPS1), a rate-limiting enzyme of pyrimidine synthesis, as a negative regulator of interferon induction. Mechanistically, CTPS1 interacts with and deamidates interferon regulatory factor 3 (IRF3). Deamidation at N85 impairs IRF3 binding to promoters containing IRF3-responsive elements, thus muting interferon (IFN) induction. Employing CTPS1 conditional deletion and IRF3 deamidated or deamidation-resistant knockin mice, we demonstrated that CTPS1-driven IRF3 deamidation restricts IFN induction in response to viral infection in vivo. However, during immune activation, IRF3 deamidation by CTPS1 is inhibited by glycogen synthase kinase 3 beta (GSK3β) to promote IFN induction. This work demonstrates how CTPS1 tames innate immunity independent of its role in pyrimidine synthesis, thus expanding the functional repertoire of metabolic enzymes into immune regulation.
Collapse
Affiliation(s)
- Youliang Rao
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Ali Can Savas
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Qizhi Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Shu Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Guoli Hou
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Taolin Xie
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
5
|
Raghav R, Awata J, Martin GL, Strathdee D, Blanton RM, Chin MT. Cell-Penetrating Peptide Enhances Tafazzin Gene Therapy in Mouse Model of Barth Syndrome. Int J Mol Sci 2024; 25:13560. [PMID: 39769321 PMCID: PMC11678443 DOI: 10.3390/ijms252413560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Barth Syndrome (BTHS) is an early onset, lethal X-linked disorder caused by a mutation in tafazzin (TAFAZZIN), a mitochondrial acyltransferase that remodels monolysocardiolipin (MLCL) to mature cardiolipin (CL) and is essential for normal mitochondrial, cardiac, and skeletal muscle function. Current gene therapies in preclinical development require high levels of transduction. We tested whether TAFAZZIN gene therapy could be enhanced with the addition of a cell-penetrating peptide, penetratin (Antp). We found that TAFAZZIN-Antp was more effective than TAFAZZIN at preventing the development of pathological cardiac hypertrophy and heart failure. These findings indicate that a cell-penetrating peptide enhances gene therapy for BTHS.
Collapse
Affiliation(s)
- Rahul Raghav
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (R.R.); (J.A.); (G.L.M.); (R.M.B.)
| | - Junya Awata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (R.R.); (J.A.); (G.L.M.); (R.M.B.)
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (R.R.); (J.A.); (G.L.M.); (R.M.B.)
| | | | - Robert M. Blanton
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (R.R.); (J.A.); (G.L.M.); (R.M.B.)
| | - Michael T. Chin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA; (R.R.); (J.A.); (G.L.M.); (R.M.B.)
| |
Collapse
|
6
|
Yue F, Ku AT, Stevens PD, Michalski MN, Jiang W, Tu J, Shi Z, Dou Y, Wang Y, Feng XH, Hostetter G, Wu X, Huang S, Shroyer NF, Zhang B, Williams BO, Liu Q, Lin X, Li Y. Loss of ZNRF3/RNF43 Unleashes EGFR in Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574969. [PMID: 38260423 PMCID: PMC10802575 DOI: 10.1101/2024.01.10.574969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
ZNRF3 and RNF43 are closely related transmembrane E3 ubiquitin ligases with significant roles in development and cancer. Conventionally, their biological functions have been associated with regulating WNT signaling receptor ubiquitination and degradation. However, our proteogenomic studies have revealed EGFR as the protein most negatively correlated with ZNRF3/RNF43 mRNA levels in multiple human cancers. Through biochemical investigations, we demonstrate that ZNRF3/RNF43 interact with EGFR via their extracellular domains, leading to EGFR ubiquitination and subsequent degradation facilitated by the E3 ligase RING domain. Overexpression of ZNRF3 reduces EGFR levels and suppresses cancer cell growth in vitro and in vivo, whereas knockout of ZNRF3/RNF43 stimulates cell growth and tumorigenesis through upregulated EGFR signaling. Together, these data highlight ZNRF3 and RNF43 as novel E3 ubiquitin ligases of EGFR and establish the inactivation of ZNRF3/RNF43 as a driver of increased EGFR signaling, ultimately promoting cancer progression. This discovery establishes a connection between two fundamental signaling pathways, EGFR and WNT, at the level of cytoplasmic membrane receptors, uncovering a novel mechanism underlying the frequent co-activation of EGFR and WNT signaling in development and cancer.
Collapse
Affiliation(s)
- Fei Yue
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Amy T. Ku
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Payton D. Stevens
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
- Biological Sciences Department, Miami University, Oxford, Ohio, 45056, USA
| | - Megan N. Michalski
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
| | - Weiyu Jiang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jianghua Tu
- Texas Therapeutics Institute and Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Zhongcheng Shi
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Galen Hostetter
- Van Andel Institute, Core Technologies and Services, Grand Rapids, Michigan 49503, USA
| | - Xiangwei Wu
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shixia Huang
- Advanced Technology Cores, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Education, Innovation & Technology, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Noah F. Shroyer
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Bart O. Williams
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA
- Van Andel Institute, Core Technologies and Services, Grand Rapids, Michigan 49503, USA
| | - Qingyun Liu
- Texas Therapeutics Institute and Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Xia Lin
- The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yi Li
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
7
|
Šimunić E, Podgorski II, Pinterić M, Hadžija MP, Belužić R, Paradžik M, Dončević L, Balog T, Kaloper M, Habisch H, Madl T, Korać A, Sobočanec S. Sirtuin 3 drives sex-specific responses to age-related changes in mouse embryonic fibroblasts. Mech Ageing Dev 2024; 222:111996. [PMID: 39395563 DOI: 10.1016/j.mad.2024.111996] [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: 07/22/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
The aging process is a complex phenomenon characterised by a gradual decline in physiological functions and an increased susceptibility to age-related diseases. An important factor in aging is mitochondrial dysfunction, which leads to an accumulation of cellular damage over time. Mitochondrial Sirtuin 3 (Sirt3), an important regulator of energy metabolism, plays a central role in maintaining mitochondrial function. Loss of Sirt3 can lead to reduced energy levels and an impaired ability to repair cellular damage, a hallmark of the aging process. In this study we investigated the impact of Sirt3 loss on mitochondrial function, metabolic responses and cellular aging processes in male and female mouse embryonic fibroblasts (MEF) exposed to etoposide-induced DNA damage, which is commonly associated with cellular dysfunction and senescence. We found that Sirt3 contributes to the sex-specific metabolic response to etoposide treatment. While male MEF exhibited minimal damage suggesting potential prior adaptation to stress due to Sirt3 loss, female MEF lacking Sirt3 experienced higher vulnerability to genotoxic stress, implying a pivotal role of Sirt3 in their resistance to such challenges. These findings offer potential insights into therapeutic strategies targeting Sirt3- and sex-specific signalling pathways in diseases associated with DNA damage that play a critical role in the aging process.
Collapse
Affiliation(s)
- Ena Šimunić
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Iva I Podgorski
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Marija Pinterić
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Marijana Popović Hadžija
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Robert Belužić
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Mladen Paradžik
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Lucija Dončević
- Department of Chemistry, Faculty of Science, University of Zagreb, Horvatovac 102a, Zagreb 10 000, Croatia.
| | - Tihomir Balog
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| | - Marta Kaloper
- Division of Molecular Biology, Faculty of Science, University of Zagreb, Ravnice 48, Zagreb 10 000, Croatia.
| | - Hansjörg Habisch
- Division of Medicinal Chemistry, Medical University of Graz, Neue Stiftingtalstraße 6, Graz 8010, Austria.
| | - Tobias Madl
- BioTechMed Graz, Mozartgasse 12/II, Graz 8010, Austria.
| | - Aleksandra Korać
- Faculty of Biology, University of Belgrade, Studentski trg 16, Beograd 11158, Serbia.
| | - Sandra Sobočanec
- Division for Molecular Medicine, Rudjer Boskovic Institute, Bijenička cesta 54, Zagreb 10 000, Croatia.
| |
Collapse
|
8
|
Zhang BQ, Wang FQ, Yin J, Yu XT, Hu ZX, Gu LH, Tong QY, Zhang YH. Michael Acceptor Pyrrolidone Derivatives and Their Activity against Diffuse Large B-cell Lymphoma. Curr Med Sci 2024; 44:890-901. [PMID: 39285051 DOI: 10.1007/s11596-024-2922-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/16/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE This study aimed to design and evaluate the efficacy of pyrrolidone derivatives as potential therapeutic agents against diffuse large B-cell lymphoma (DLBCL), a common and heterogeneous malignancy of the adult lymphohematopoietic system. Given the limitations of current therapies, there is a pressing need to develop new and effective drugs for DLBCL treatment. METHODS A series of pyrrolidone derivatives were synthesized, and their antitumor activities were assessed, particularly against DLBCL cell lines. Structure-activity relationship (SAR) analysis was conducted to identify key structural components essential for activity. The most promising compound, referred to as compound 7, was selected for further mechanistic studies. The expression levels of relevant mRNA and protein were detected by RT-qPCR and Western blotting, and the expression of mitochondrial membrane potential and ROS was detected using flow cytometry for further assessment of cell cycle arrest and apoptosis. RESULTS The compound 7 exhibited good antitumor activity among the synthesized derivatives, specifically in DLBCL cell lines. SAR analysis highlighted the critical role of α, β-unsaturated ketones in the antitumor efficacy of these compounds. Mechanistically, compound 7 was found to induce significant DNA damage, trigger an inflammatory response, cause mitochondrial dysfunction, and disrupt cell cycle progression, ultimately leading to apoptosis of DLBCL cells. CONCLUSION The compound 7 has good antitumor activity and can induce multiple cellular mechanisms leading to cancer cell death. These findings warrant further investigation of the compound 7 as a potential therapeutic agent for DLBCL.
Collapse
Affiliation(s)
- Bi-Qiong Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng-Qing Wang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Yin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Tan Yu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zheng-Xi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liang-Hu Gu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Qing-Yi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yong-Hui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
9
|
Aquino LVCD, Olindo SL, Silva YLFE, Oliveira LRMD, Moura YBF, Rodrigues ALR, Praxedes ÉA, Oliveira MFD, Silva AR, Pereira AF. Cryopreservation and passaging optimization for Galea spixii (Wagler, 1831) adult skin fibroblast lines: A step forward in species management and genetic studies. Acta Histochem 2024; 126:152185. [PMID: 39059228 DOI: 10.1016/j.acthis.2024.152185] [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: 03/22/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND In vitro culture of fibroblasts is a technique based on cell isolation, physiological characterization, and cryopreservation. This technique has not been described for Galea spixii, therefore, it can be used to learn about its cellular biology and genetic diversity. OBJECTIVE We established fibroblast lines of six G. spixii individuals from several passages (second, fifth, eighth, and tenth) and cryopreserved them. METHODS Fibroblasts recovered from skin biopsies were identified based on morphology, immunocytochemistry, and karyotyping. The cells were analyzed for morphology, ultrastructure, viability, proliferation, metabolism, oxidative stress, bioenergetic potential, and apoptosis before and after cryopreservation. RESULTS After the eighth passage, the fibroblasts showed morphological and karyotypic changes, although their viability, metabolism, and proliferation did not change. An increase in oxidative stress and bioenergetic potential from the fifth to the eighth passages were also observed. Post cryopreservation, cell damage with respect to the ultrastructure, viability, proliferative rate, apoptotic levels, oxidative stress, and bioenergetic potential were verified. CONCLUSION Fibroblasts up to the tenth passage could be cultured in vitro. However, cells at the fifth passage were of better quality to be used for reproductive techniques. Additionally, optimization of the cryopreservation protocol is essential to improve the physiological parameters of these cells.
Collapse
Affiliation(s)
| | - Samara Lima Olindo
- Laboratory of Animal Biotechnology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | | | | | | | | | - Érika Almeida Praxedes
- Laboratory of Animal Biotechnology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil. erikaalmeida-@hotmail.com
| | - Moacir Franco de Oliveira
- Laboratory of Applied Animal Morphophysiology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | - Alexandre Rodrigues Silva
- Laboratory of Animal Germplasma Conservation, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | | |
Collapse
|
10
|
Cormerais Y, Lapp SC, Kalafut KC, Cissé MY, Shin J, Stefadu B, Personnaz J, Schrotter S, D’Amore A, Martin ER, Salussolia CL, Sahin M, Menon S, Byles V, Manning BD. AKT-mediated phosphorylation of TSC2 controls stimulus- and tissue-specific mTORC1 signaling and organ growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614519. [PMID: 39386441 PMCID: PMC11463511 DOI: 10.1101/2024.09.23.614519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) integrates diverse intracellular and extracellular growth signals to regulate cell and tissue growth. How the molecular mechanisms regulating mTORC1 signaling established through biochemical and cell biological studies function under physiological states in specific mammalian tissues are unknown. Here, we characterize a genetic mouse model lacking the 5 phosphorylation sites on the tuberous sclerosis complex 2 (TSC2) protein through which the growth factor-stimulated protein kinase AKT can active mTORC1 signaling in cell culture models. These phospho-mutant mice (TSC2-5A) are developmentally normal but exhibit reduced body weight and the weight of specific organs, such as brain and skeletal muscle, associated with cell intrinsic decreases in growth factor-stimulated mTORC1 signaling. The TSC2-5A mouse model demonstrates that TSC2 phosphorylation is a primary mechanism of mTORC1 activation in some, but not all, tissues and provides a genetic tool to facilitate studies on the physiological regulation of mTORC1.
Collapse
Affiliation(s)
- Yann Cormerais
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Samuel C. Lapp
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Krystle C. Kalafut
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Madi Y. Cissé
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Jong Shin
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Benjamin Stefadu
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jean Personnaz
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Sandra Schrotter
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Cell Signaling Technologies, Inc, Beverly, MA, 01915, USA
| | - Angelica D’Amore
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Emma R. Martin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Catherine L. Salussolia
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Suchithra Menon
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Vanessa Byles
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Brendan D. Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
11
|
Prashar A, Bussi C, Fearns A, Capurro MI, Gao X, Sesaki H, Gutierrez MG, Jones NL. Lysosomes drive the piecemeal removal of mitochondrial inner membrane. Nature 2024; 632:1110-1117. [PMID: 39169179 PMCID: PMC7616637 DOI: 10.1038/s41586-024-07835-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Mitochondrial membranes define distinct structural and functional compartments. Cristae of the inner mitochondrial membrane (IMM) function as independent bioenergetic units that undergo rapid and transient remodelling, but the significance of this compartmentalized organization is unknown1. Using super-resolution microscopy, here we show that cytosolic IMM vesicles, devoid of outer mitochondrial membrane or mitochondrial matrix, are formed during resting state. These vesicles derived from the IMM (VDIMs) are formed by IMM herniation through pores formed by voltage-dependent anion channel 1 in the outer mitochondrial membrane. Live-cell imaging showed that lysosomes in proximity to mitochondria engulfed the herniating IMM and, aided by the endosomal sorting complex required for transport machinery, led to the formation of VDIMs in a microautophagy-like process, sparing the remainder of the organelle. VDIM formation was enhanced in mitochondria undergoing oxidative stress, suggesting their potential role in maintenance of mitochondrial function. Furthermore, the formation of VDIMs required calcium release by the reactive oxygen species-activated, lysosomal calcium channel, transient receptor potential mucolipin 1, showing an interorganelle communication pathway for maintenance of mitochondrial homeostasis. Thus, IMM compartmentalization could allow for the selective removal of damaged IMM sections via VDIMs, which should protect mitochondria from localized injury. Our findings show a new pathway of intramitochondrial quality control.
Collapse
Affiliation(s)
- Akriti Prashar
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- NHLBI, NIH, Bethesda, MD, USA
| | - Claudio Bussi
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
- School of Biological Sciences, Nanyang Technical University, Singapore, Singapore
| | - Antony Fearns
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Mariana I Capurro
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaodong Gao
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, UK
| | - Nicola L Jones
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Limerick A, McCabe EA, Turner JS, Kuang KW, Brautigan DL, Hao Y, Chu CY, Fu SH, Ahmadi S, Xu W, Fu Z. An Epilepsy-Associated CILK1 Variant Compromises KATNIP Regulation and Impairs Primary Cilia and Hedgehog Signaling. Cells 2024; 13:1258. [PMID: 39120290 PMCID: PMC11311665 DOI: 10.3390/cells13151258] [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: 05/30/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Mutations in human CILK1 (ciliogenesis associated kinase 1) are linked to ciliopathies and epilepsy. Homozygous point and nonsense mutations that extinguish kinase activity impair primary cilia function, whereas mutations outside the kinase domain are not well understood. Here, we produced a knock-in mouse equivalent to the human CILK1 A615T variant identified in juvenile myoclonic epilepsy (JME). This residue is in the intrinsically disordered C-terminal region of CILK1 separate from the kinase domain. Mouse embryo fibroblasts (MEFs) with either heterozygous or homozygous A612T mutant alleles exhibited a higher ciliation rate, shorter individual cilia, and upregulation of ciliary Hedgehog signaling. Thus, a single A612T mutant allele was sufficient to impair primary cilia and ciliary signaling in MEFs. Gene expression profiles of wild-type versus mutant MEFs revealed profound changes in cilia-related molecular functions and biological processes. The CILK1 A615T mutant protein was not increased to the same level as the wild-type protein when co-expressed with scaffold protein KATNIP (katanin-interacting protein). Our data show that KATNIP regulation of a JME-associated single-residue variant of CILK1 is compromised, and this impairs the maintenance of primary cilia and Hedgehog signaling.
Collapse
Affiliation(s)
- Ana Limerick
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Ellie A. McCabe
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Jacob S. Turner
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Kevin W. Kuang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - David L. Brautigan
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (D.L.B.); (W.X.)
| | - Yi Hao
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA;
| | - Cheuk Ying Chu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Sean H. Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Sean Ahmadi
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Wenhao Xu
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (D.L.B.); (W.X.)
| | - Zheng Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| |
Collapse
|
13
|
Li Y, Li L, Wang X, Zhao F, Yang Y, Zhou Y, Zhang J, Wang L, Jiang Z, Zhang Y, Chen Y, Wu C, Li K, Zhang T, Wang P, Mao Z, Zhu W, Xu X, Liang S, Lou Z, Yuan J. USP25 Elevates SHLD2-Mediated DNA Double-Strand Break Repair and Regulates Chemoresponse in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403485. [PMID: 38803048 PMCID: PMC11267380 DOI: 10.1002/advs.202403485] [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: 04/03/2024] [Indexed: 05/29/2024]
Abstract
DNA damage plays a significant role in the tumorigenesis and progression of the disease. Abnormal DNA repair affects the therapy and prognosis of cancer. In this study, it is demonstrated that the deubiquitinase USP25 promotes non-homologous end joining (NHEJ), which in turn contributes to chemoresistance in cancer. It is shown that USP25 deubiquitinates SHLD2 at the K64 site, which enhances its binding with REV7 and promotes NHEJ. Furthermore, USP25 deficiency impairs NHEJ-mediated DNA repair and reduces class switch recombination (CSR) in USP25-deficient mice. USP25 is overexpressed in a subset of colon cancers. Depletion of USP25 sensitizes colon cancer cells to IR, 5-Fu, and cisplatin. TRIM25 is also identified, an E3 ligase, as the enzyme responsible for degrading USP25. Downregulation of TRIM25 leads to an increase in USP25 levels, which in turn induces chemoresistance in colon cancer cells. Finally, a peptide that disrupts the USP25-SHLD2 interaction is successfully identified, impairing NHEJ and increasing sensitivity to chemotherapy in PDX model. Overall, these findings reveal USP25 as a critical effector of SHLD2 in regulating the NHEJ repair pathway and suggest its potential as a therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Yunhui Li
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Cancer CenterTongji University School of MedicineShanghai200331China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Lei Li
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Cancer CenterTongji University School of MedicineShanghai200331China
| | - Xinshu Wang
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Fei Zhao
- College of BiologyHunan UniversityChangsha410082China
| | - Yuntong Yang
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Yujuan Zhou
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Jiyuan Zhang
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Li Wang
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Zeshan Jiang
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| | - Yuanyuan Zhang
- Department of General Surgery and Colorectal SurgeryShanghai East HospitalTongji University School of MedicineShanghai200120China
| | - Yuping Chen
- Cancer CenterTongji University School of MedicineShanghai200331China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
- Translational Research Institute of Brain and Brain‐Like IntelligenceShanghai Fourth People's HospitalSchool of MedicineTongji UniversityShanghai200080China
| | - Chenming Wu
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Cancer CenterTongji University School of MedicineShanghai200331China
| | - Ke Li
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Medicinal BiotechnologyChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Tingting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Medicinal BiotechnologyChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100050China
| | - Ping Wang
- Tongji University Cancer CenterShanghai Tenth People's HospitalSchool of MedicineShanghai200072China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal‐Fetal MedicineClinical and Translational Research Center of Shanghai First Maternity and Infant HospitalFrontier Science Center for Stem Cell ResearchTongji University School of MedicineShanghai200040China
| | - Weiguo Zhu
- International Cancer CenterGuangdong Key Laboratory of Genome Instability and Human Disease PreventionMarshall Laboratory of Biomedical EngineeringDepartment of Biochemistry and Molecular BiologyShenzhen University Medical SchoolShenzhen518037China
| | - Xingzhi Xu
- The Sixth Affiliated Hospital of Shenzhen UniversityGuangdong Key Laboratory for Genome Stability and Disease Prevention and Carson International Cancer CenterMarshall Laboratory of Biomedical EngineeringShenzhen University School of MedicineShenzhen518055China
| | - Shikang Liang
- School of Biomedical SciencesLKS Faculty of MedicineThe University of Hong KongHong Kong SAR999077Hong Kong
| | - Zhenkun Lou
- Department of OncologyMayo ClinicRochesterMNUSA
| | - Jian Yuan
- Medical Innovation CenterShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
- Cancer CenterTongji University School of MedicineShanghai200331China
- Department of Biochemistry and Molecular BiologyTongji University School of MedicineShanghai200331China
| |
Collapse
|
14
|
Kagi T, Inoue A, Noguchi T, Suzuki W, Takano S, Otani K, Naganuma R, Sekiguchi Y, Hirata Y, Shindo S, Hwang GW, Matsuzawa A. The NLRP3 Inflammasome Is a Major Cause of Acute Renal Failure Induced by Polypeptide Antibiotics. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1807-1818. [PMID: 38639584 DOI: 10.4049/jimmunol.2300193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 03/16/2024] [Indexed: 04/20/2024]
Abstract
Drug-induced acute renal failure (ARF) is a public health concern that hinders optimal drug therapy. However, pathological mechanisms of drug-induced ARF remain to be elucidated. Here, we show that a pathological process of drug-induced ARF is mediated by proinflammatory cross-talk between kidney tubular cells and macrophages. Both polymyxin B and colistin, polypeptide antibiotics, frequently cause ARF, stimulated the ERK and NF-κB pathways in kidney tubular cells, and thereby upregulated M-CSF and MCP-1, leading to infiltration of macrophages into the kidneys. Thereafter, the kidney-infiltrated macrophages were exposed to polypeptide antibiotics, which initiated activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. Interestingly, blockade of the NLRP3 activation clearly ameliorated the pathology of ARF induced by polypeptide antibiotics, suggesting that a combination of the distinct cellular responses to polypeptide antibiotics in kidney tubular cells and macrophages plays a key role in the pathogenesis of colistin-induced ARF. Thus, our results provide a concrete example of how drugs initiate ARF, which may give insight into the underlying pathological process of drug-induced ARF.
Collapse
Affiliation(s)
- Tomohiro Kagi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Aya Inoue
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Wakana Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Saya Takano
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohei Otani
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Rio Naganuma
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yuto Sekiguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sawako Shindo
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Environmental Toxicology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
15
|
Vanhoutte D, Schips TG, Minerath RA, Huo J, Kavuri NSS, Prasad V, Lin SC, Bround MJ, Sargent MA, Adams CM, Molkentin JD. Thbs1 regulates skeletal muscle mass in a TGFβ-Smad2/3-ATF4-dependent manner. Cell Rep 2024; 43:114149. [PMID: 38678560 PMCID: PMC11217783 DOI: 10.1016/j.celrep.2024.114149] [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: 08/08/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/01/2024] Open
Abstract
Loss of muscle mass is a feature of chronic illness and aging. Here, we report that skeletal muscle-specific thrombospondin-1 transgenic mice (Thbs1 Tg) have profound muscle atrophy with age-dependent decreases in exercise capacity and premature lethality. Mechanistically, Thbs1 activates transforming growth factor β (TGFβ)-Smad2/3 signaling, which also induces activating transcription factor 4 (ATF4) expression that together modulates the autophagy-lysosomal pathway (ALP) and ubiquitin-proteasome system (UPS) to facilitate muscle atrophy. Indeed, myofiber-specific inhibition of TGFβ-receptor signaling represses the induction of ATF4, normalizes ALP and UPS, and partially restores muscle mass in Thbs1 Tg mice. Similarly, myofiber-specific deletion of Smad2 and Smad3 or the Atf4 gene antagonizes Thbs1-induced muscle atrophy. More importantly, Thbs1-/- mice show significantly reduced levels of denervation- and caloric restriction-mediated muscle atrophy, along with blunted TGFβ-Smad3-ATF4 signaling. Thus, Thbs1-mediated TGFβ-Smad3-ATF4 signaling in skeletal muscle regulates tissue rarefaction, suggesting a target for atrophy-based muscle diseases and sarcopenia with aging.
Collapse
Affiliation(s)
- Davy Vanhoutte
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tobias G Schips
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachel A Minerath
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jiuzhou Huo
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Naga Swathi Sree Kavuri
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Suh-Chin Lin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael J Bround
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle A Sargent
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Christopher M Adams
- Division of Endocrinology, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
16
|
Sikder K, Phillips E, Zhong Z, Wang N, Saunders J, Mothy D, Kossenkov A, Schneider T, Nichtova Z, Csordas G, Margulies KB, Choi JC. Perinuclear damage from nuclear envelope deterioration elicits stress responses that contribute to LMNA cardiomyopathy. SCIENCE ADVANCES 2024; 10:eadh0798. [PMID: 38718107 PMCID: PMC11078192 DOI: 10.1126/sciadv.adh0798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2024] [Indexed: 05/12/2024]
Abstract
Mutations in the LMNA gene encoding lamins A/C cause an array of tissue-selective diseases, with the heart being the most commonly affected organ. Despite progress in understanding the perturbations emanating from LMNA mutations, an integrative understanding of the pathogenesis underlying cardiac dysfunction remains elusive. Using a novel conditional deletion model capable of translatome profiling, we observed that cardiomyocyte-specific Lmna deletion in adult mice led to rapid cardiomyopathy with pathological remodeling. Before cardiac dysfunction, Lmna-deleted cardiomyocytes displayed nuclear abnormalities, Golgi dilation/fragmentation, and CREB3-mediated stress activation. Translatome profiling identified MED25 activation, a transcriptional cofactor that regulates Golgi stress. Autophagy is disrupted in the hearts of these mice, which can be recapitulated by disrupting the Golgi. Systemic administration of modulators of autophagy or ER stress significantly delayed cardiac dysfunction and prolonged survival. These studies support a hypothesis wherein stress responses emanating from the perinuclear space contribute to the LMNA cardiomyopathy development.
Collapse
Affiliation(s)
- Kunal Sikder
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Elizabeth Phillips
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Zhijiu Zhong
- Translational Research and Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nadan Wang
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Jasmine Saunders
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - David Mothy
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| | - Andrew Kossenkov
- Bioinformatics Facility, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Timothy Schneider
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zuzana Nichtova
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gyorgy Csordas
- Mitocare, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kenneth B. Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia PA, USA
| |
Collapse
|
17
|
Georgieva D, Wang N, Taglialatela A, Jerabek S, Reczek CR, Lim PX, Sung J, Du Q, Horiguchi M, Jasin M, Ciccia A, Baer R, Egli D. BRCA1 and 53BP1 regulate reprogramming efficiency by mediating DNA repair pathway choice at replication-associated double-strand breaks. Cell Rep 2024; 43:114006. [PMID: 38554279 PMCID: PMC11272184 DOI: 10.1016/j.celrep.2024.114006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 11/26/2023] [Accepted: 03/11/2024] [Indexed: 04/01/2024] Open
Abstract
Reprogramming to pluripotency is associated with DNA damage and requires the functions of the BRCA1 tumor suppressor. Here, we leverage separation-of-function mutations in BRCA1/2 as well as the physical and/or genetic interactions between BRCA1 and its associated repair proteins to ascertain the relevance of homology-directed repair (HDR), stalled fork protection (SFP), and replication gap suppression (RGS) in somatic cell reprogramming. Surprisingly, loss of SFP and RGS is inconsequential for the transition to pluripotency. In contrast, cells deficient in HDR, but proficient in SFP and RGS, reprogram with reduced efficiency. Conversely, the restoration of HDR function through inactivation of 53bp1 rescues reprogramming in Brca1-deficient cells, and 53bp1 loss leads to elevated HDR and enhanced reprogramming in mouse and human cells. These results demonstrate that somatic cell reprogramming is especially dependent on repair of replication-associated double-strand breaks (DSBs) by the HDR activity of BRCA1 and BRCA2 and can be improved in the absence of 53BP1.
Collapse
Affiliation(s)
- Daniela Georgieva
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia University Stem Cell Initiative, New York, NY 10032, USA
| | - Ning Wang
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia University Stem Cell Initiative, New York, NY 10032, USA
| | - Angelo Taglialatela
- Columbia University Stem Cell Initiative, New York, NY 10032, USA; Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Stepan Jerabek
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia University Stem Cell Initiative, New York, NY 10032, USA; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 160 00 Praha 6, Czech Republic
| | - Colleen R Reczek
- Department of Pathology & Cell Biology, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pei Xin Lim
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julie Sung
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Qian Du
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michiko Horiguchi
- Department of Pathology & Cell Biology, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Maria Jasin
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alberto Ciccia
- Columbia University Stem Cell Initiative, New York, NY 10032, USA; Department of Genetics and Development, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Richard Baer
- Department of Pathology & Cell Biology, Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dieter Egli
- Department of Pediatrics and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia University Stem Cell Initiative, New York, NY 10032, USA; Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
18
|
Belužić R, Šimunić E, Podgorski II, Pinterić M, Hadžija MP, Balog T, Sobočanec S. Gene Expression Profiling Reveals Fundamental Sex-Specific Differences in SIRT3-Mediated Redox and Metabolic Signaling in Mouse Embryonic Fibroblasts. Int J Mol Sci 2024; 25:3868. [PMID: 38612678 PMCID: PMC11012119 DOI: 10.3390/ijms25073868] [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: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Sirt-3 is an important regulator of mitochondrial function and cellular energy homeostasis, whose function is associated with aging and various pathologies such as Alzheimer's disease, Parkinson's disease, cardiovascular diseases, and cancers. Many of these conditions show differences in incidence, onset, and progression between the sexes. In search of hormone-independent, sex-specific roles of Sirt-3, we performed mRNA sequencing in male and female Sirt-3 WT and KO mouse embryonic fibroblasts (MEFs). The aim of this study was to investigate the sex-specific cellular responses to the loss of Sirt-3. By comparing WT and KO MEF of both sexes, the differences in global gene expression patterns as well as in metabolic and stress responses associated with the loss of Sirt-3 have been elucidated. Significant differences in the activities of basal metabolic pathways were found both between genotypes and between sexes. In-depth pathway analysis of metabolic pathways revealed several important sex-specific phenomena. Male cells mount an adaptive Hif-1a response, shifting their metabolism toward glycolysis and energy production from fatty acids. Furthermore, the loss of Sirt-3 in male MEFs leads to mitochondrial and endoplasmic reticulum stress. Since Sirt-3 knock-out is permanent, male cells are forced to function in a state of persistent oxidative and metabolic stress. Female MEFs are able to at least partially compensate for the loss of Sirt-3 by a higher expression of antioxidant enzymes. The activation of neither Hif-1a, mitochondrial stress response, nor oxidative stress response was observed in female cells lacking Sirt-3. These findings emphasize the sex-specific role of Sirt-3, which should be considered in future research.
Collapse
|
19
|
Lauterboeck L, Kang SW, White D, Bao R, Mobasheran P, Yang Q. IF1 Promotes Cellular Proliferation and Inhibits Oxidative Phosphorylation in Mouse Embryonic Fibroblasts under Normoxia and Hypoxia. Cells 2024; 13:551. [PMID: 38534395 PMCID: PMC10969582 DOI: 10.3390/cells13060551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
ATP synthase inhibitory factor subunit 1 (IF1) is an inhibitory subunit of mitochondrial ATP synthase, playing a crucial role in regulating mitochondrial respiration and energetics. It is well-established that IF1 interacts with the F1 sector of ATP synthase to inhibit the reversal rotation and, thus, ATP hydrolysis. Recent evidence supports that IF1 also inhibits forward rotation or the ATP synthesis activity. Adding to the complexity, IF1 may also facilitate mitophagy and cristae formation. The implications of these complex actions of IF1 for cellular function remain obscure. In the present study, we found that IF1 expression was markedly upregulated in hypoxic MEFs relative to normoxic MEFs. We investigate how IF1 affects cellular growth and function in cultured mouse embryonic fibroblasts derived from mouse lines with systemic IF1 overexpression and knockout under normoxia and hypoxia. Cell survival and proliferation analyses revealed that IF1 overexpression exerted limited effects on cellular viability but substantially increased proliferation under normoxia, whereas it facilitated both cellular viability and proliferation under hypoxia. The absence of IF1 may have a pro-survival effect but not a proliferative one in both normoxia and hypoxia. Cellular bioenergetic analyses revealed that IF1 suppressed cellular respiration when subjected to normoxia and was even more pronounced when subjected to hypoxia with increased mitochondrial ATP production. In contrast, IF1 knockout MEFs showed markedly increased cellular respiration under both normoxia and hypoxia with little change in mitochondrial ATP. Glycolytic stress assay revealed that IF1 overexpression modestly increased glycolysis in normoxia and hypoxia. Interestingly, the absence of IF1 in MEFs led to substantial increases in glycolysis. Therefore, we conclude that IF1 mainly inhibits cellular respiration and enhances cellular glycolysis to preserve mitochondrial ATP. On the other hand, IF1 deletion can significantly facilitate cellular respiration and glycolysis without leading to mitochondrial ATP deficit.
Collapse
Affiliation(s)
- Lothar Lauterboeck
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Cell Biology, Life Science Solutions, Thermo Fisher Scientific, Frederick, MD 21704, USA
| | - Sung Wook Kang
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
| | - Donnell White
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Rong Bao
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Parnia Mobasheran
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Qinglin Yang
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (L.L.); (S.W.K.); (D.W.III); (R.B.); (P.M.)
- Department of Pharmacology and Experimental Therapeutics, School of Graduate Studies, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
20
|
Maharaj AV, Cottrell E, Thanasupawat T, Joustra SD, Triggs-Raine B, Fujimoto M, Kant SG, van der Kaay D, Clement-de Boers A, Brooks AS, Aguirre GA, Martín del Estal I, Castilla de Cortázar Larrea MI, Massoud A, van Duyvenvoorde HA, De Bruin C, Hwa V, Klonisch T, Hombach-Klonisch S, Storr HL. Characterization of HMGA2 variants expands the spectrum of Silver-Russell syndrome. JCI Insight 2024; 9:e169425. [PMID: 38516887 PMCID: PMC11063932 DOI: 10.1172/jci.insight.169425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Silver-Russell syndrome (SRS) is a heterogeneous disorder characterized by intrauterine and postnatal growth retardation. HMGA2 variants are a rare cause of SRS and its functional role in human linear growth is unclear. Patients with suspected SRS negative for 11p15LOM/mUPD7 underwent whole-exome and/or targeted-genome sequencing. Mutant HMGA2 protein expression and nuclear localization were assessed. Two Hmga2-knockin mouse models were generated. Five clinical SRS patients harbored HMGA2 variants with differing functional impacts: 2 stop-gain nonsense variants (c.49G>T, c.52C>T), c.166A>G missense variant, and 2 frameshift variants (c.144delC, c.145delA) leading to an identical, extended-length protein. Phenotypic features were highly variable. Nuclear localization was reduced/absent for all variants except c.166A>G. Homozygous knockin mice recapitulating the c.166A>G variant (Hmga2K56E) exhibited a growth-restricted phenotype. An Hmga2Ter76-knockin mouse model lacked detectable full-length Hmga2 protein, similarly to patient 3 and 5 variants. These mice were infertile, with a pygmy phenotype. We report a heterogeneous group of individuals with SRS harboring variants in HMGA2 and describe the first Hmga2 missense knockin mouse model (Hmga2K56E) to our knowledge causing a growth-restricted phenotype. In patients with clinical features of SRS but negative genetic screening, HMGA2 should be included in next-generation sequencing testing approaches.
Collapse
Affiliation(s)
- Avinaash V. Maharaj
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Emily Cottrell
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Thatchawan Thanasupawat
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sjoerd D. Joustra
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Barbara Triggs-Raine
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Masanobu Fujimoto
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Sarina G. Kant
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Danielle van der Kaay
- Division of Paediatric Endocrinology, Department of Paediatrics, Erasmus University Medical Centre, Sophia Children’s Hospital, Rotterdam, Netherlands
| | - Agnes Clement-de Boers
- Department of Paediatrics, Juliana Children’s Hospital/Haga Teaching Hospital, The Hague, Netherlands
| | - Alice S. Brooks
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | | | | | | | - Ahmed Massoud
- Department of Paediatrics and Child Health, HCA Healthcare UK, London, United Kingdom
| | - Hermine A. van Duyvenvoorde
- Laboratory for Diagnostic Genome analysis (LDGA), Department of Clinical Genetics, Leiden University Medical Centre, Leiden, Netherlands
| | - Christiaan De Bruin
- Division of Paediatric Endocrinology, Department of Paediatrics, Willem-Alexander Children’s Hospital, Leiden University Medical Centre, Leiden, Netherlands
| | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pathology, and
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pathology, and
| | - Helen L. Storr
- Centre for Endocrinology, William Harvey Research Institute, QMUL, London, United Kingdom
| |
Collapse
|
21
|
Li Z, Xu T, Li X, Wang T, Tang G, Zhao H, Zhao Y, Ye K, Gao P. Viral integration promotes SV40T-induced immortalization by disturbing the expression of DNA/chromosome- and ECM-associated functional genes. Gene 2024; 896:148060. [PMID: 38048968 DOI: 10.1016/j.gene.2023.148060] [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: 10/18/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/06/2023]
Abstract
Lentivirus containing simian virus 40 large T antigen (SV40T) is routinely used to induce cell immortalization. However, the roles of viral integration itself in this progress is still controversial. Here, we transformed primary mouse embryonic fibroblasts (MEFs) with SV40T lentivirus and studied the roles of viral integration in the immortalization using RNA sequencing (RNA-seq) and whole genome sequencing (WGS). During the immortalization, differentially expressed genes (DGEs) are enriched in viral infection and several diverse activities. However, DEGs between immortalized and aging cells are significantly enriched in DNA/chromosome- and extracellular matrix (ECM)-associated activities. Gene regulatory network (GRN) analysis shows that although p53 is a key regulatory factor, many other transcription factors also play critical roles in the process, like STAT1. Of these DEGs, 32 genes have viral integration in their coding and/or regulatory regions. Our findings suggest that viral integration may promote SV40T-mediated immortalization by disturbing the expression of DNA/chromosome- and ECM-associated genes.
Collapse
Affiliation(s)
- Zihang Li
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tun Xu
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xiujuan Li
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tingjie Wang
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guangbo Tang
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Huanhuan Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yuanli Zhao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kai Ye
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an 710049, China; School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China; Faculty of Science, Leiden University, Leiden, the Netherlands.
| | - Peng Gao
- Genome Institute, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
22
|
Lin YH, Lehle JD, McCarrey JR. Source cell-type epigenetic memory persists in induced pluripotent cells but is lost in subsequently derived germline cells. Front Cell Dev Biol 2024; 12:1306530. [PMID: 38410371 PMCID: PMC10895008 DOI: 10.3389/fcell.2024.1306530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction: Retention of source cell-type epigenetic memory may mitigate the potential for induced pluripotent stem cells (iPSCs) to fully achieve transitions in cell fate in vitro. While this may not preclude the use of iPSC-derived somatic cell types for therapeutic applications, it becomes a major concern impacting the potential use of iPSC-derived germline cell types for reproductive applications. The transition from a source somatic cell type to iPSCs and then on to germ-cell like cells (GCLCs) recapitulates two major epigenetic reprogramming events that normally occur during development in vivo-embryonic reprogramming in the epiblast and germline reprogramming in primordial germ cells (PGCs). We examined the extent of epigenetic and transcriptomic memory persisting first during the transition from differentiated source cell types to iPSCs, and then during the transition from iPSCs to PGC-like cells (PGCLCs). Methods: We derived iPSCs from four differentiated mouse cell types including two somatic and two germ cell types and tested the extent to which each resulting iPSC line resembled a) a validated ES cell reference line, and b) their respective source cell types, on the basis of genome-wide gene expression and DNA methylation patterns. We then induced each iPSC line to form PGCLCs, and assessed epigenomic and transcriptomic memory in each compared to endogenous PGCs/M-prospermatogonia. Results: In each iPSC line, we found residual gene expression and epigenetic programming patterns characteristic of the corresponding source differentiated cell type from which each was derived. However, upon deriving PGCLCs, we found very little evidence of lingering epigenetic or transcriptomic memory of the original source cell type. Discussion: This result indicates that derivation of iPSCs and then GCLCs from differentiated source cell types in vitro recapitulates the two-phase epigenetic reprogramming that normally occurs in vivo, and that, to a significant extent, germline cell types derived in vitro from pluripotent cells accurately recapitulate epigenetic programming and gene expression patterns corresponding to equivalent endogenous germ cell types, suggesting that they have the potential to form the basis of in vitro gametogenesis as a useful therapeutic strategy for treatment of infertility.
Collapse
Affiliation(s)
- Yu-Huey Lin
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jake D Lehle
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| | - John R McCarrey
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
23
|
Higuchi Y, Ogata T, Nakanishi N, Nishi M, Tsuji Y, Tomita S, Conway SJ, Matoba S. Cavin-2 promotes fibroblast-to-myofibroblast trans-differentiation and aggravates cardiac fibrosis. ESC Heart Fail 2024; 11:167-178. [PMID: 37872863 PMCID: PMC10804157 DOI: 10.1002/ehf2.14571] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023] Open
Abstract
AIMS Transforming growth factor β (TGF-β) signalling is one of the critical pathways in fibroblast activation, and several drugs targeting the TGF-β/Smad signalling pathway in heart failure with cardiac fibrosis are being tested in clinical trials. Some caveolins and cavins, which are components of caveolae on the plasma membrane, are known for their association with the regulation of TGF-β signalling. Cavin-2 is particularly abundant in fibroblasts; however, the detailed association between Cavin-2 and cardiac fibrosis is still unclear. We tried to clarify the involvement and role of Cavin-2 in fibroblasts and cardiac fibrosis. METHODS AND RESULTS To clarify the role of Cavin-2 in cardiac fibrosis, we performed transverse aortic constriction (TAC) operations on four types of mice: wild-type (WT), Cavin-2 null (Cavin-2 KO), Cavin-2flox/flox , and activated fibroblast-specific Cavin-2 conditional knockout (Postn-Cre/Cavin-2flox/flox , Cavin-2 cKO) mice. We collected mouse embryonic fibroblasts (MEFs) from WT and Cavin-2 KO mice and investigated the effect of Cavin-2 in fibroblast trans-differentiation into myofibroblasts and associated TGF-β signalling. Four weeks after TAC, cardiac fibrotic areas in both the Cavin-2 KO and the Cavin-2 cKO mice were significantly decreased compared with each control group (WT 8.04 ± 1.58% vs. Cavin-2 KO 0.40 ± 0.03%, P < 0.01; Cavin-2flox/flox , 7.19 ± 0.50% vs. Cavin-2 cKO 0.88 ± 0.44%, P < 0.01). Fibrosis-associated mRNA expression (Col1a1, Ctgf, and Col3) was significantly attenuated in the Cavin-2 KO mice after TAC. α1 type I collagen deposition and non-vascular αSMA-positive cells (WT 43.5 ± 2.4% vs. Cavin-2 KO 25.4 ± 3.2%, P < 0.01) were reduced in the heart of the Cavin-2 cKO mice after TAC operation. The levels of αSMA protein (0.36-fold, P < 0.05) and fibrosis-associated mRNA expression (Col1a1, 0.69-fold, P < 0.01; Ctgf, 0.27-fold, P < 0.01; Col3, 0.60-fold, P < 0.01) were decreased in the Cavin-2 KO MEFs compared with the WT MEFs. On the other hand, αSMA protein levels were higher in the Cavin-2 overexpressed MEFs compared with the control MEFs (2.40-fold, P < 0.01). TGF-β1-induced Smad2 phosphorylation was attenuated in the Cavin-2 KO MEFs compared with WT MEFs (0.60-fold, P < 0.01). Heat shock protein 90 protein levels were significantly reduced in the Cavin-2 KO MEFs compared with the WT MEFs (0.69-fold, P < 0.01). CONCLUSIONS Cavin-2 loss suppressed fibroblast trans-differentiation into myofibroblasts through the TGF-β/Smad signalling. The loss of Cavin-2 in cardiac fibroblasts suppresses cardiac fibrosis and may maintain cardiac function.
Collapse
Affiliation(s)
- Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pathology and Cell Regulation, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Naohiko Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Department of PediatricsIndiana University School of MedicineIndianapolisINUSA
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
24
|
Liu K, Zhao C, Adajar RC, DeZwaan-McCabe D, Rutkowski DT. A beneficial adaptive role for CHOP in driving cell fate selection during ER stress. EMBO Rep 2024; 25:228-253. [PMID: 38177915 PMCID: PMC10897205 DOI: 10.1038/s44319-023-00026-0] [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: 03/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 01/06/2024] Open
Abstract
Cellular stresses elicit signaling cascades that are capable of either mitigating the inciting dysfunction or initiating cell death. During endoplasmic reticulum (ER) stress, the transcription factor CHOP is widely recognized to promote cell death. However, it is not clear whether CHOP also has a beneficial role during adaptation. Here, we combine a new, versatile, genetically modified Chop allele with single cell analysis and with stresses of physiological intensity, to rigorously examine the contribution of CHOP to cell fate. Paradoxically, we find that CHOP promotes death in some cells, but proliferation-and hence recovery-in others. Strikingly, this function of CHOP confers to cells a stress-specific competitive growth advantage. The dynamics of CHOP expression and UPR activation at the single cell level suggest that CHOP maximizes UPR activation, which in turn favors stress resolution, subsequent UPR deactivation, and proliferation. Taken together, these findings suggest that CHOP's function can be better described as a "stress test" that drives cells into either of two mutually exclusive fates-adaptation or death-during stresses of physiological intensity.
Collapse
Affiliation(s)
- Kaihua Liu
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Chaoxian Zhao
- Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Reed C Adajar
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Diane DeZwaan-McCabe
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - D Thomas Rutkowski
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
25
|
Chen Z, Kwan SY, Mir A, Hazeltine M, Shin M, Liang SQ, Chan IL, Kelly K, Ghanta KS, Gaston N, Cao Y, Xie J, Gao G, Xue W, Sontheimer EJ, Watts JK. A Fluorescent Reporter Mouse for In Vivo Assessment of Genome Editing with Diverse Cas Nucleases and Prime Editors. CRISPR J 2023; 6:570-582. [PMID: 38108517 PMCID: PMC10753986 DOI: 10.1089/crispr.2023.0048] [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: 07/27/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
CRISPR-based genome-editing technologies, including nuclease editing, base editing, and prime editing, have recently revolutionized the development of therapeutics targeting disease-causing mutations. To advance the assessment and development of genome editing tools, a robust mouse model is valuable, particularly for evaluating in vivo activity and delivery strategies. In this study, we successfully generated a knock-in mouse line carrying the Traffic Light Reporter design known as TLR-multi-Cas variant 1 (TLR-MCV1). We comprehensively validated the functionality of this mouse model for both in vitro and in vivo nuclease and prime editing. The TLR-MCV1 reporter mouse represents a versatile and powerful tool for expediting the development of editing technologies and their therapeutic applications.
Collapse
Affiliation(s)
- Zexiang Chen
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Suet-Yan Kwan
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Aamir Mir
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Max Hazeltine
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Minwook Shin
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Shun-Qing Liang
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Io Long Chan
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Karen Kelly
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Krishna S. Ghanta
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas Gaston
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Yueying Cao
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jun Xie
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Viral Vector Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Wen Xue
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Erik J. Sontheimer
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jonathan K. Watts
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- NeuroNexus Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
26
|
Maus M, López-Polo V, Mateo L, Lafarga M, Aguilera M, De Lama E, Meyer K, Sola A, Lopez-Martinez C, López-Alonso I, Guasch-Piqueras M, Hernandez-Gonzalez F, Chaib S, Rovira M, Sanchez M, Faner R, Agusti A, Diéguez-Hurtado R, Ortega S, Manonelles A, Engelhardt S, Monteiro F, Stephan-Otto Attolini C, Prats N, Albaiceta G, Cruzado JM, Serrano M. Iron accumulation drives fibrosis, senescence and the senescence-associated secretory phenotype. Nat Metab 2023; 5:2111-2130. [PMID: 38097808 PMCID: PMC10730403 DOI: 10.1038/s42255-023-00928-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Fibrogenesis is part of a normal protective response to tissue injury that can become irreversible and progressive, leading to fatal diseases. Senescent cells are a main driver of fibrotic diseases through their secretome, known as senescence-associated secretory phenotype (SASP). Here, we report that cellular senescence, and multiple types of fibrotic diseases in mice and humans are characterized by the accumulation of iron. We show that vascular and hemolytic injuries are efficient in triggering iron accumulation, which in turn can cause senescence and promote fibrosis. Notably, we find that senescent cells persistently accumulate iron, even when the surge of extracellular iron has subdued. Indeed, under normal conditions of extracellular iron, cells exposed to different types of senescence-inducing insults accumulate abundant ferritin-bound iron, mostly within lysosomes, and present high levels of labile iron, which fuels the generation of reactive oxygen species and the SASP. Finally, we demonstrate that detection of iron by magnetic resonance imaging might allow non-invasive assessment of fibrotic burden in the kidneys of mice and in patients with renal fibrosis. Our findings suggest that iron accumulation plays a central role in senescence and fibrosis, even when the initiating events may be independent of iron, and identify iron metabolism as a potential therapeutic target for senescence-associated diseases.
Collapse
Grants
- SAF2017-82613-R "la Caixa" Foundation (Caixa Foundation)
- of M. Serrano was funded by the IRB and “laCaixa” Foundation, and by grants from the Spanish Ministry of Science co-funded by the European Regional Development Fund (ERDF) (SAF2017-82613-R), European Research Council (ERC-2014-AdG/669622), and grant RETOS COLABORACION RTC2019-007125-1 from MCIN/AEI, and Secretaria d'Universitats i Recerca del Departament d'Empresa i Coneixement of Catalonia (Grup de Recerca consolidat 2017 SGR 282)
- M.M. received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement (No 794744) and from the Spanish Ministry of Science and Innovation (MCIN) (RYC2020-030652-I /AEI /10.13039/501100011033)
- V.L.P. was recipient of a predoctoral contract from Spanish Ministry of Education (FPU-18/05917).
- K.M. was recipient of fellowships from the German Cardiac, the German Research Foundation, and a postdoctoral contract Juan de la Cierva from the MCIN.
- F.H.G. was supported by the PhD4MD Collaborative Research Training Programme for Medical Doctors (IRB Barcelona/Hospital Clinic/IDIBAPS).
- M. Sanchez was funded by grants PID2021-122436OB-I00 from MCIN/ AEI /10.13039/501100011033 / FEDER, UE, and RETOS COLABORACION RTC2019-007074-1 from MCIN/AEI /10.13039/501100011033.
- G.A. was funded by Instituto de Salud Carlos III through project PI 20/01360, FEDER funds.
- J.M.C was funded by Instituto de Salud Carlos III through projects PI18/00910 and PI21/00931 (Co-funded by European Regional Development Fund. ERDF, a way to build Europe), and thanks CERCA Programme / Generalitat de Catalunya for institutional support.
Collapse
Affiliation(s)
- Mate Maus
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | - Vanessa López-Polo
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Lidia Mateo
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Miguel Lafarga
- Departamento de Anatomía y Biología Celular, Universidad de Cantabria-IDIVAL, Santander, Spain
| | - Mònica Aguilera
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eugenia De Lama
- Radiology Department, Hospital Universitari de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Kathleen Meyer
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Anna Sola
- Nephrology and Renal Transplantation Research Group. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Cecilia Lopez-Martinez
- Departamento de Biología Funcional, Instituto Universitario de Oncología del principado de Asturias, Universidad de Oviedo, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos. Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- CIBER-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Ines López-Alonso
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | | | - Fernanda Hernandez-Gonzalez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Pulmonary Medicine, Respiratory Institute, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Selim Chaib
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Miguel Rovira
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Mayka Sanchez
- Iron Metabolism: Regulation and Diseases Group, Department of Basic Sciences, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès, Spain
| | - Rosa Faner
- Biomedicine Department, University of Barcelona, IDIBAPS, CIBERES, Barcelona, Spain
| | - Alvar Agusti
- Universitat de Barcelona, Institut Respiratori, Hospital Clinic, IDIBAPS, CIBERES, Barcelona, Spain
| | - Rodrigo Diéguez-Hurtado
- Deparment of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Sagrario Ortega
- Transgenics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Anna Manonelles
- Nephrology and Renal Transplantation Research Group. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Freddy Monteiro
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Guillermo Albaiceta
- Departamento de Biología Funcional, Instituto Universitario de Oncología del principado de Asturias, Universidad de Oviedo, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos. Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- CIBER-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep M Cruzado
- Nephrology and Renal Transplantation Research Group. Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
- Nephrology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
- Department of Clinical Sciences, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Altos Labs, Cambridge Institute of Science, Cambridge, UK.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
27
|
Auroni TT, Arora K, Natekar JP, Pathak H, Elsharkawy A, Kumar M. The critical role of interleukin-6 in protection against neurotropic flavivirus infection. Front Cell Infect Microbiol 2023; 13:1275823. [PMID: 38053527 PMCID: PMC10694511 DOI: 10.3389/fcimb.2023.1275823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/27/2023] [Indexed: 12/07/2023] Open
Abstract
West Nile virus (WNV) and Japanese encephalitis virus (JEV) are emerging mosquito-borne flaviviruses causing encephalitis globally. No specific drug or therapy exists to treat flavivirus-induced neurological diseases. The lack of specific therapeutics underscores an urgent need to determine the function of important host factors involved in flavivirus replication and disease progression. Interleukin-6 (IL-6) upregulation has been observed during viral infections in both mice and humans, implying that it may influence the disease outcome significantly. Herein, we investigated the function of IL-6 in the pathogenesis of neurotropic flavivirus infections. First, we examined the role of IL-6 in flavivirus-infected human neuroblastoma cells, SK-N-SH, and found that IL-6 neutralization increased the WNV or JEV replication and inhibited the expression of key cytokines. We further evaluated the role of IL-6 by infecting primary mouse cells derived from IL-6 knockout (IL-6-/-) mice and wild-type (WT) mice with WNV or JEV. The results exhibited increased virus yields in the cells lacking the IL-6 gene. Next, our in vivo approach revealed that IL-6-/- mice had significantly higher morbidity and mortality after subcutaneous infection with the pathogenic WNV NY99 or JEV Nakayama strain compared to WT mice. The non-pathogenic WNV Eg101 strain did not cause mortality in WT mice but resulted in 60% mortality in IL-6-/- mice, indicating that IL-6 is required for the survival of mice after the peripheral inoculation of WNV or JEV. We also observed significantly higher viremia and brain viral load in IL-6-/- mice than in WT mice. Subsequently, we explored innate immune responses in WT and IL-6-/- mice after WNV NY99 infection. Our data demonstrated that the IL-6-/- mice had reduced levels of key cytokines in the serum during early infection but elevated levels of proinflammatory cytokines in the brain later, along with suppressed anti-inflammatory cytokines. In addition, mRNA expression of IFN-α and IFN-β was significantly lower in the infected IL-6-/- mice. In conclusion, these data suggest that the lack of IL-6 exacerbates WNV or JEV infection in vitro and in vivo by causing an increase in virus replication and dysregulating host immune response.
Collapse
Affiliation(s)
| | | | | | | | | | - Mukesh Kumar
- Department of Biology, College of Arts and Sciences, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
28
|
Liu K, Zhao C, Adajar RC, DeZwaan-McCabe D, Rutkowski DT. A beneficial adaptive role for CHOP in driving cell fate selection during ER stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533325. [PMID: 36993175 PMCID: PMC10055232 DOI: 10.1101/2023.03.19.533325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cellular stresses elicit signaling cascades that are capable of either mitigating the inciting dysfunction or initiating cell death. During endoplasmic reticulum (ER) stress, the transcription factor CHOP is widely recognized to promote cell death. However, it is not clear whether CHOP also has a beneficial role during adaptation. Here, we have combined a new, versatile, genetically modified Chop allele with single cell analysis and with stresses of physiological intensity, to rigorously examine the contribution of CHOP to cell fate. Paradoxically, we found that CHOP promoted death in some cells, but proliferation-and hence recovery-in others. Strikingly, this function of CHOP conferred to cells a stress-specific competitive growth advantage. The dynamics of CHOP expression and UPR activation at the single cell level suggested that CHOP maximizes UPR activation, which in turn favors stress resolution, subsequent UPR deactivation, and proliferation. Taken together, these findings suggest that CHOP's function can be better described as a "stress test" that drives cells into either of two mutually exclusive fates-adaptation or death-during stresses of physiological intensity.
Collapse
Affiliation(s)
- Kaihua Liu
- Program in Human Toxicology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Chaoxian Zhao
- Shanghai Cancer Institute, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Reed C. Adajar
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Diane DeZwaan-McCabe
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - D. Thomas Rutkowski
- Program in Human Toxicology, University of Iowa Carver College of Medicine, Iowa City, IA
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA
| |
Collapse
|
29
|
Golotin V, Lyutikov A, Filatova T, Sharoyko V, Apalikova O. A Rapid and Simple Procedure for the Isolation of Embryonic Cells from Fish Eggs. Bio Protoc 2023; 13:e4836. [PMID: 37817904 PMCID: PMC10560692 DOI: 10.21769/bioprotoc.4836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 10/12/2023] Open
Abstract
Fertilized teleost fish eggs are a complex formation, in which dividing cells are located in a small point in the entire volume of eggs. Isolating embryonic cells can be considered a necessary step in the research of developmental peculiarities of fish cells at the earliest stages of embryogenesis before embryo formation. The main advantages of the offered protocol are rapid isolation, no enzymes, and overall low cost compared to other protocols. The protocol is suitable for the isolation of embryonic cells from medium-sized eggs at the stages of blastula or gastrula, for studies in a variety of applications (e.g., microscopy, flow cytometry, and other methods). Fertilized nelma eggs (Stenodus leucichthys nelma) are used in the protocol as a model. Key features • Fast and cheap isolation of cells from fish eggs at early stages (blastula or gastrula). • Applicable for most of the methods for cell study (any staining, microscopy, flow cytometry, etc.). • Can be applied to other teleost fish eggs with medium egg diameter of 3-4 mm. Graphical overview.
Collapse
Affiliation(s)
- Vasily Golotin
- Saint Petersburg branch of “VNIRO”, “GosNIORH” named after L.S. Berg, Saint-Petersburg, Russia
- First Saint-Petersburg State Medical University, Saint-Petersburg, Russia
| | - Anatoly Lyutikov
- Saint Petersburg branch of “VNIRO”, “GosNIORH” named after L.S. Berg, Saint-Petersburg, Russia
| | - Tatiana Filatova
- Saint Petersburg branch of “VNIRO”, “GosNIORH” named after L.S. Berg, Saint-Petersburg, Russia
| | - Vladimir Sharoyko
- First Saint-Petersburg State Medical University, Saint-Petersburg, Russia
| | - Olga Apalikova
- Saint Petersburg branch of “VNIRO”, “GosNIORH” named after L.S. Berg, Saint-Petersburg, Russia
| |
Collapse
|
30
|
Mooring M, Yeung GA, Luukkonen P, Liu S, Akbar MW, Zhang GJ, Balogun O, Yu X, Mo R, Nejak-Bowen K, Poyurovsky MV, Booth CJ, Konnikova L, Shulman GI, Yimlamai D. Hepatocyte CYR61 polarizes profibrotic macrophages to orchestrate NASH fibrosis. Sci Transl Med 2023; 15:eade3157. [PMID: 37756381 PMCID: PMC10874639 DOI: 10.1126/scitranslmed.ade3157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Obesity is increasing worldwide and leads to a multitude of metabolic diseases, including cardiovascular disease, type 2 diabetes, nonalcoholic fatty liver disease, and nonalcoholic steatohepatitis (NASH). Cysteine-rich angiogenic inducer 61 (CYR61) is associated with the progression of NASH, but it has been described to have anti- and proinflammatory properties. We sought to examine the role of liver CYR61 in NASH progression. CYR61 liver-specific knockout mice on a NASH diet showed improved glucose tolerance, decreased liver inflammation, and reduced fibrosis. CYR61 polarized infiltrating monocytes promoting a proinflammatory/profibrotic phenotype through an IRAK4/SYK/NF-κB signaling cascade. In vitro, CYR61 activated a profibrotic program, including PDGFa/PDGFb expression in macrophages, in an IRAK4/SYK/NF-κB-dependent manner. Furthermore, targeted-antibody blockade reduced CYR61-driven signaling in macrophages in vitro and in vivo, reducing fibrotic development. This study demonstrates that CYR61 is a key driver of liver inflammation and fibrosis in NASH.
Collapse
Affiliation(s)
- Meghan Mooring
- Department of Cellular and Molecular Pathology, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
- These authors contributed equally to this work
| | - Grace A. Yeung
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
- These authors contributed equally to this work
| | - Panu Luukkonen
- Department of Internal Medicine, Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Silvia Liu
- Department of Pathology, School of Medicine, University of Pittsburgh
- Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
| | - Muhammad Waqas Akbar
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Gary J. Zhang
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Oluwashanu Balogun
- Department of Cellular and Molecular Pathology, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
- Department of Pathology, School of Medicine, University of Pittsburgh
| | - Xuemei Yu
- Kadmon Corporation, LLC; 450 East 29th Street, New York, New York 10016, USA
| | - Rigen Mo
- Kadmon Corporation, LLC; 450 East 29th Street, New York, New York 10016, USA
| | - Kari Nejak-Bowen
- Department of Cellular and Molecular Pathology, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
- Department of Pathology, School of Medicine, University of Pittsburgh
- Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
| | - Masha V. Poyurovsky
- Kadmon Corporation, LLC; 450 East 29th Street, New York, New York 10016, USA
| | - Carmen J. Booth
- Department of Comparative Medicine; Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Liza Konnikova
- Section of Neonatology; Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale School of Medicine; New Haven, Connecticut 06514, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine; New Haven, Connecticut 06514, USA
| | - Dean Yimlamai
- Department of Cellular and Molecular Pathology, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics; Yale School of Medicine; New Haven, Connecticut 06514, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine; Pittsburgh, Pennsylvania 15261, USA
- The Yale Liver Center, Yale School of Medicine; New Haven, Connecticut 06514, USA
| |
Collapse
|
31
|
Leppert HG, Anderson JT, Timm KJ, Davoli C, Pratt MA, Booth CD, White KA, Rechtzigel MJ, Meyerink BL, Johnson TB, Brudvig JJ, Weimer JM. Sortilin inhibition treats multiple neurodegenerative lysosomal storage disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.559064. [PMID: 37790379 PMCID: PMC10543011 DOI: 10.1101/2023.09.22.559064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Lysosomal storage disorders (LSDs) are a genetically and clinically diverse group of diseases characterized by lysosomal dysfunction. Batten disease is a family of severe LSDs primarily impacting the central nervous system. Here we show that AF38469, a small molecule inhibitor of sortilin, improves lysosomal and glial pathology across multiple LSD models. Live-cell imaging and comparative transcriptomics demonstrates that the transcription factor EB (TFEB), an upstream regulator of lysosomal biogenesis, is activated upon treatment with AF38469. Utilizing CLN2 and CLN3 Batten disease mouse models, we performed a short-term efficacy study and show that treatment with AF38469 prevents the accumulation of lysosomal storage material and the development of neuroinflammation, key disease associated pathologies. Tremor phenotypes, an early behavioral phenotype in the CLN2 disease model, were also completely rescued. These findings reveal sortilin inhibition as a novel and highly efficacious therapeutic modality for the treatment of multiple forms of Batten disease.
Collapse
Affiliation(s)
- Hannah G. Leppert
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | | | - Kaylie J. Timm
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | - Cristina Davoli
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | - Melissa A. Pratt
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | - Clarissa D. Booth
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | | | | | | | - Tyler B. Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
| | - Jon J. Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| |
Collapse
|
32
|
Bao Y, Oh JH, Kang CW, Ku CR, Cho YH, Lee EJ. Olfactory marker protein regulates adipogenesis via the cAMP-IκBα pathway. Mol Cell Endocrinol 2023; 575:111992. [PMID: 37328092 DOI: 10.1016/j.mce.2023.111992] [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: 05/04/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/18/2023]
Abstract
Olfactory marker protein (OMP) regulates olfactory transduction and is also expressed in adipose tissue. Since it serves as a regulatory buffer for cyclic AMP (cAMP) levels, we hypothesized that it plays a role in modulating adipocyte differentiation. To determine the role of OMP in adipogenesis, we examined the differences in body weight, adipose tissue mass, and adipogenic or thermogenic gene expression between high-fat diet-fed control and Omp-knockout (KO) mice. cAMP production, adipogenic gene expression, and cAMP response element binding protein (CREB) phosphorylation were measured during the differentiation of 3T3-L1 preadipocytes and mouse embryonic fibroblasts (MEFs). RNA sequencing was performed to determine the gene expression patterns responsible for the reduction in adipogenesis when Omp was deleted. Body weight, adipose tissue mass, and adipocyte size decreased in Omp-KO mice. Furthermore, cAMP production and CREB phosphorylation reduced during adipogenesis induced in Omp-/- MEFs, and the Nuclear factor kappa B was activated due to significantly reduced expression of its inhibitor. Collectively, our results suggest that loss of OMP function inhibits adipogenesis by affecting adipocyte differentiation.
Collapse
Affiliation(s)
- Yaru Bao
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University, College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ju Hun Oh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chan Woo Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Cheol Ryong Ku
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yoon Hee Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Eun Jig Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University, College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
33
|
Martinez-Lopez N, Mattar P, Toledo M, Bains H, Kalyani M, Aoun ML, Sharma M, McIntire LBJ, Gunther-Cummins L, Macaluso FP, Aguilan JT, Sidoli S, Bourdenx M, Singh R. mTORC2-NDRG1-CDC42 axis couples fasting to mitochondrial fission. Nat Cell Biol 2023:10.1038/s41556-023-01163-3. [PMID: 37386153 PMCID: PMC10344787 DOI: 10.1038/s41556-023-01163-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 05/04/2023] [Indexed: 07/01/2023]
Abstract
Fasting triggers diverse physiological adaptations including increases in circulating fatty acids and mitochondrial respiration to facilitate organismal survival. The mechanisms driving mitochondrial adaptations and respiratory sufficiency during fasting remain incompletely understood. Here we show that fasting or lipid availability stimulates mTORC2 activity. Activation of mTORC2 and phosphorylation of its downstream target NDRG1 at serine 336 sustains mitochondrial fission and respiratory sufficiency. Time-lapse imaging shows that NDRG1, but not the phosphorylation-deficient NDRG1Ser336Ala mutant, engages with mitochondria to facilitate fission in control cells, as well as in those lacking DRP1. Using proteomics, a small interfering RNA screen, and epistasis experiments, we show that mTORC2-phosphorylated NDRG1 cooperates with small GTPase CDC42 and effectors and regulators of CDC42 to orchestrate fission. Accordingly, RictorKO, NDRG1Ser336Ala mutants and Cdc42-deficient cells each display mitochondrial phenotypes reminiscent of fission failure. During nutrient surplus, mTOR complexes perform anabolic functions; however, paradoxical reactivation of mTORC2 during fasting unexpectedly drives mitochondrial fission and respiration.
Collapse
Affiliation(s)
- Nuria Martinez-Lopez
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Liver Basic Research Center at University of California Los Angeles, Los Angeles, CA, USA
| | - Pamela Mattar
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Miriam Toledo
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Henrietta Bains
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Manu Kalyani
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marie Louise Aoun
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mridul Sharma
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Leslie Gunther-Cummins
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Frank P Macaluso
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jennifer T Aguilan
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mathieu Bourdenx
- UK Dementia Research Institute, London, UK
- UCL Queen Square Institute of Neurology, London, UK
| | - Rajat Singh
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
- Liver Basic Research Center at University of California Los Angeles, Los Angeles, CA, USA.
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
34
|
Xu AF, Molinuevo R, Fazzari E, Tom H, Zhang Z, Menendez J, Casey KM, Ruggero D, Hinck L, Pritchard JK, Barna M. Subfunctionalized expression drives evolutionary retention of ribosomal protein paralogs Rps27 and Rps27l in vertebrates. eLife 2023; 12:e78695. [PMID: 37306301 PMCID: PMC10313321 DOI: 10.7554/elife.78695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/09/2023] [Indexed: 06/13/2023] Open
Abstract
The formation of paralogs through gene duplication is a core evolutionary process. For paralogs that encode components of protein complexes such as the ribosome, a central question is whether they encode functionally distinct proteins or whether they exist to maintain appropriate total expression of equivalent proteins. Here, we systematically tested evolutionary models of paralog function using the ribosomal protein paralogs Rps27 (eS27) and Rps27l (eS27L) as a case study. Evolutionary analysis suggests that Rps27 and Rps27l likely arose during whole-genome duplication(s) in a common vertebrate ancestor. We show that Rps27 and Rps27l have inversely correlated mRNA abundance across mouse cell types, with the highest Rps27 in lymphocytes and the highest Rps27l in mammary alveolar cells and hepatocytes. By endogenously tagging the Rps27 and Rps27l proteins, we demonstrate that Rps27- and Rps27l-ribosomes associate preferentially with different transcripts. Furthermore, murine Rps27 and Rps27l loss-of-function alleles are homozygous lethal at different developmental stages. However, strikingly, expressing Rps27 protein from the endogenous Rps27l locus or vice versa completely rescues loss-of-function lethality and yields mice with no detectable deficits. Together, these findings suggest that Rps27 and Rps27l are evolutionarily retained because their subfunctionalized expression patterns render both genes necessary to achieve the requisite total expression of two equivalent proteins across cell types. Our work represents the most in-depth characterization of a mammalian ribosomal protein paralog to date and highlights the importance of considering both protein function and expression when investigating paralogs.
Collapse
Affiliation(s)
- Adele Francis Xu
- Department of Genetics, Stanford UniversityStanfordUnited States
- Medical Scientist Training Program, Stanford School of MedicineStanfordUnited States
| | - Rut Molinuevo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa CruzSanta CruzUnited States
| | - Elisa Fazzari
- Helen Diller Family Comprehensive Cancer Center, University of California, Los AngelesLos AngelesUnited States
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
| | - Harrison Tom
- Helen Diller Family Comprehensive Cancer Center, University of California, Los AngelesLos AngelesUnited States
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
| | - Zijian Zhang
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Julien Menendez
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa CruzSanta CruzUnited States
| | - Kerriann M Casey
- Department of Biology, Stanford UniversityStanfordUnited States
- Department of Comparative Medicine, Stanford School of MedicineStanfordUnited States
| | - Davide Ruggero
- Department of Cellular and Molecular Pharmacology, University of California, San FranciscoSan FranciscoUnited States
- Department of Urology, University of California, San FranciscoSan FranciscoUnited States
| | - Lindsay Hinck
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa CruzSanta CruzUnited States
| | | | - Maria Barna
- Department of Genetics, Stanford UniversityStanfordUnited States
| |
Collapse
|
35
|
Nguyen DD, Kim E, Le NT, Ding X, Jaiswal RK, Kostlan RJ, Nguyen TNT, Shiva O, Le MT, Chai W. Deficiency in mammalian STN1 promotes colon cancer development via inhibiting DNA repair. SCIENCE ADVANCES 2023; 9:eadd8023. [PMID: 37163605 PMCID: PMC10171824 DOI: 10.1126/sciadv.add8023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/05/2023] [Indexed: 05/12/2023]
Abstract
Despite the high lethality of colorectal cancers (CRCs), only a limited number of genetic risk factors are identified. The mammalian ssDNA-binding protein complex CTC1-STN1-TEN1 protects genome stability, yet its role in tumorigenesis is unknown. Here, we show that attenuated CTC1/STN1 expression is common in CRCs. We generated an inducible STN1 knockout mouse model and found that STN1 deficiency in young adult mice increased CRC incidence, tumor size, and tumor load. CRC tumors exhibited enhanced proliferation, reduced apoptosis, and elevated DNA damage and replication stress. We found that STN1 deficiency down-regulated multiple DNA glycosylases, resulting in defective base excision repair (BER) and accumulation of oxidative damage. Collectively, this study identifies STN1 deficiency as a risk factor for CRC and implicates the previously unknown STN1-BER axis in protecting colon tissues from oxidative damage, therefore providing insights into the CRC tumor-suppressing mechanism.
Collapse
Affiliation(s)
- Dinh Duc Nguyen
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Eugene Kim
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Nhat Thong Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Rishi Kumar Jaiswal
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Raymond Joseph Kostlan
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Thi Ngoc Thanh Nguyen
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Olga Shiva
- Office of Research, Washington State University-Spokane, Spokane, WA, USA
| | - Minh Thong Le
- School of Biotechnology, International University, Ho Chi Minh City, Vietnam
| | - Weihang Chai
- Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
36
|
Seong J, Rivron NC. Protocol for capturing trophectoderm stem cells reflecting the blastocyst stage. STAR Protoc 2023; 4:102151. [PMID: 36930647 PMCID: PMC10031533 DOI: 10.1016/j.xpro.2023.102151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/13/2022] [Accepted: 02/10/2023] [Indexed: 03/18/2023] Open
Abstract
Classically, culturing mouse blastocysts with FGF4/TGF-β1, two epiblast-secreted inducers, allows for deriving trophoblast stem cells that comprise fluctuating subpopulations reflecting both pre- and post-implantation stages. However, a more complete combination of inducers (adding LPA, IL11, BMP7, Activin A, 8-Br-cAMP) captures trophectoderm stem cells with enhanced transcriptomic similarity to the blastocyst trophectoderm and self-renewal, reduced differentiation. Also, the complete combination of inducers increased potential to form blastoids and to instruct decidualization in utero, thus better reflecting the blastocyst. For complete details on the use and execution of this protocol, please refer to Seong et al.1.
Collapse
Affiliation(s)
- Jinwoo Seong
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| | - Nicolas C Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria.
| |
Collapse
|
37
|
Vom Stein AF, Rebollido-Rios R, Lukas A, Koch M, von Lom A, Reinartz S, Bachurski D, Rose F, Bozek K, Abdallah AT, Kohlhas V, Saggau J, Zölzer R, Zhao Y, Bruns C, Bröckelmann PJ, Lohneis P, Büttner R, Häupl B, Oellerich T, Nguyen PH, Hallek M. LYN kinase programs stromal fibroblasts to facilitate leukemic survival via regulation of c-JUN and THBS1. Nat Commun 2023; 14:1330. [PMID: 36899005 PMCID: PMC10006233 DOI: 10.1038/s41467-023-36824-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 02/14/2023] [Indexed: 03/12/2023] Open
Abstract
Microenvironmental bystander cells are essential for the progression of chronic lymphocytic leukemia (CLL). We have discovered previously that LYN kinase promotes the formation of a microenvironmental niche for CLL. Here we provide mechanistic evidence that LYN regulates the polarization of stromal fibroblasts to support leukemic progression. LYN is overexpressed in fibroblasts of lymph nodes of CLL patients. LYN-deficient stromal cells reduce CLL growth in vivo. LYN-deficient fibroblasts show markedly reduced leukemia feeding capacity in vitro. Multi-omics profiling reveals that LYN regulates the polarization of fibroblasts towards an inflammatory cancer-associated phenotype through modulation of cytokine secretion and extracellular matrix composition. Mechanistically, LYN deletion reduces inflammatory signaling including reduction of c-JUN expression, which in turn augments the expression of Thrombospondin-1, which binds to CD47 thereby impairing CLL viability. Together, our findings suggest that LYN is essential for rewiring fibroblasts towards a leukemia-supportive phenotype.
Collapse
Affiliation(s)
- Alexander F Vom Stein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Rocio Rebollido-Rios
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Anna Lukas
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Maximilian Koch
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Anton von Lom
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Sebastian Reinartz
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Daniel Bachurski
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - France Rose
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- University of Cologne, Institute for Biomedical Informatics, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Katarzyna Bozek
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
- University of Cologne, Institute for Biomedical Informatics, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Ali T Abdallah
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Viktoria Kohlhas
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Julia Saggau
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Rebekka Zölzer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Yue Zhao
- Faculty of Medicine and University Hospital Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Christiane Bruns
- Faculty of Medicine and University Hospital Cologne, Department of General, Visceral and Cancer Surgery, University of Cologne, Cologne, Germany
| | - Paul J Bröckelmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Max-Planck Institute for the Biology of Ageing, Cologne, Germany
| | - Philipp Lohneis
- Reference Centre for Lymph Node Pathology and Hematopathology, Hämatopathologie Lübeck, Lübeck, Germany
- Faculty of Medicine and University Hospital Cologne, Department of Pathology, University of Cologne, Cologne, Germany
| | - Reinhard Büttner
- Faculty of Medicine and University Hospital Cologne, Department of Pathology, University of Cologne, Cologne, Germany
| | - Björn Häupl
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Oellerich
- Department of Hematology/Oncology, Johann Wolfgang Goethe University, Frankfurt, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Phuong-Hien Nguyen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
| | - Michael Hallek
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Cologne, Germany.
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
- CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
38
|
Ko A, Hasanain M, Oh YT, D'Angelo F, Sommer D, Frangaj B, Tran S, Bielle F, Pollo B, Paterra R, Mokhtari K, Soni RK, Peyre M, Eoli M, Papi L, Kalamarides M, Sanson M, Iavarone A, Lasorella A. LZTR1 Mutation Mediates Oncogenesis through Stabilization of EGFR and AXL. Cancer Discov 2023; 13:702-723. [PMID: 36445254 DOI: 10.1158/2159-8290.cd-22-0376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 09/23/2022] [Accepted: 11/21/2022] [Indexed: 12/02/2022]
Abstract
LZTR1 is the substrate-specific adaptor of a CUL3-dependent ubiquitin ligase frequently mutated in sporadic and syndromic cancer. We combined biochemical and genetic studies to identify LZTR1 substrates and interrogated their tumor-driving function in the context of LZTR1 loss-of-function mutations. Unbiased screens converged on EGFR and AXL receptor tyrosine kinases as LZTR1 interactors targeted for ubiquitin-dependent degradation in the lysosome. Pathogenic cancer-associated mutations of LZTR1 failed to promote EGFR and AXL degradation, resulting in dysregulated growth factor signaling. Conditional inactivation of Lztr1 and Cdkn2a in the mouse nervous system caused tumors in the peripheral nervous system including schwannoma-like tumors, thus recapitulating aspects of schwannomatosis, the prototype tumor predisposition syndrome sustained by LZTR1 germline mutations. Lztr1- and Cdkn2a-deleted tumors aberrantly accumulated EGFR and AXL and exhibited specific vulnerability to EGFR and AXL coinhibition. These findings explain tumorigenesis by LZTR1 inactivation and offer therapeutic opportunities to patients with LZTR1-mutant cancer. SIGNIFICANCE EGFR and AXL are substrates of LZTR1-CUL3 ubiquitin ligase. The frequent somatic and germline mutations of LZTR1 in human cancer cause EGFR and AXL accumulation and deregulated signaling. LZTR1-mutant tumors show vulnerability to concurrent inhibition of EGFR and AXL, thus providing precision targeting to patients affected by LZTR1-mutant cancer. This article is highlighted in the In This Issue feature, p. 517.
Collapse
Affiliation(s)
- Aram Ko
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Mohammad Hasanain
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Young Taek Oh
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Danika Sommer
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Brulinda Frangaj
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
| | - Suzanne Tran
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Laboratory of Neuropathology, Paris, France
| | - Franck Bielle
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Laboratory of Neuropathology, Paris, France
| | - Bianca Pollo
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rosina Paterra
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Karima Mokhtari
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Neurosurgery Service, Paris, France
| | - Rajesh Kumar Soni
- Proteomics Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Matthieu Peyre
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Neurosurgery Service, Paris, France
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Service of Neurology 2-Mazarin, Equipe lLNCC, Paris, France
| | - Marica Eoli
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Laura Papi
- The Department of Experimental and Clinical, Medical Genetics Unit, Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Michel Kalamarides
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Neurosurgery Service, Paris, France
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Service of Neurology 2-Mazarin, Equipe lLNCC, Paris, France
| | - Marc Sanson
- Sorbonne Université, INSERM U1127, CNRS UMR 7225, Brain Institute, ICM, AP-HP, University Hospital La Pitié Salpêtrière-Charles Foix, Service of Neurology 2-Mazarin, Equipe lLNCC, Paris, France
- Onconeurotek Tumor Bank, Brain and Spinal Cord Institute ICM, 75013 Paris, France
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
- Department of Neurology, Columbia University Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York
- Department of Pediatrics, Columbia University Medical Center, New York, New York
| |
Collapse
|
39
|
Investigation of SAMD1 ablation in mice. Sci Rep 2023; 13:3000. [PMID: 36810619 PMCID: PMC9944271 DOI: 10.1038/s41598-023-29779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/10/2023] [Indexed: 02/23/2023] Open
Abstract
SAM domain-containing protein 1 (SAMD1) has been implicated in atherosclerosis, as well as in chromatin and transcriptional regulation, suggesting a versatile and complex biological function. However, its role at an organismal level is currently unknown. Here, we generated SAMD1-/- and SAMD1+/- mice to explore the role of SAMD1 during mouse embryogenesis. Homozygous loss of SAMD1 was embryonic lethal, with no living animals seen after embryonic day 18.5. At embryonic day 14.5, organs were degrading and/or incompletely developed, and no functional blood vessels were observed, suggesting failed blood vessel maturation. Sparse red blood cells were scattered and pooled, primarily near the embryo surface. Some embryos had malformed heads and brains at embryonic day 15.5. In vitro, SAMD1 absence impaired neuronal differentiation processes. Heterozygous SAMD1 knockout mice underwent normal embryogenesis and were born alive. Postnatal genotyping showed a reduced ability of these mice to thrive, possibly due to altered steroidogenesis. In summary, the characterization of SAMD1 knockout mice suggests a critical role of SAMD1 during developmental processes in multiple organs and tissues.
Collapse
|
40
|
Schulz MS, Sartorius von Bach CB, Marinkovic E, Günther C, Behrendt R, Roers A. Development of an RNase H2 Activity Assay for Clinical Screening. J Clin Med 2023; 12:1598. [PMID: 36836134 PMCID: PMC9961991 DOI: 10.3390/jcm12041598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
As the key enzyme mediating ribonucleotide excision repair, RNase H2 is essential for the removal of single ribonucleotides from DNA in order to prevent genome damage. Loss of RNase H2 activity directly contributes to the pathogenesis of autoinflammatory and autoimmune diseases and might further play a role in ageing and neurodegeneration. Moreover, RNase H2 activity is a potential diagnostic and prognostic marker in several types of cancer. Until today, no method for quantification of RNase H2 activity has been validated for the clinical setting. Herein, validation and benchmarks of a FRET-based whole-cell lysate RNase H2 activity assay are presented, including standard conditions and procedures to calculate standardized RNase H2 activity. Spanning a wide working range, the assay is applicable to various human cell or tissue samples with overall methodological assay variability from 8.6% to 16%. Using our assay, we found RNase H2 activity was reduced in lymphocytes of two patients with systemic lupus erythematosus and one with systemic sclerosis carrying heterozygous mutations in one of the RNASEH2 genes. Implementation of larger control groups will help to assess the diagnostic and prognostic value of clinical screening for RNase H2 activity in the future.
Collapse
Affiliation(s)
- Marian Simon Schulz
- University Hospital for Children and Adolescents, University of Leipzig, 04103 Leipzig, Germany
| | | | - Emilija Marinkovic
- Institute for Immunology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Claudia Günther
- Department of Dermatology, University Hospital Carl Gustav Carus Dresden, TU Dresden, 01307 Dresden, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - Axel Roers
- Institute for Immunology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Hariri H, Kose O, Bezdjian A, Daniel SJ, St-Arnaud R. USP53 Regulates Bone Homeostasis by Controlling Rankl Expression in Osteoblasts and Bone Marrow Adipocytes. J Bone Miner Res 2023; 38:578-596. [PMID: 36726200 DOI: 10.1002/jbmr.4778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/12/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023]
Abstract
In the skeleton, osteoblasts and osteoclasts synchronize their activities to maintain bone homeostasis and integrity. Investigating the molecular mechanisms governing bone remodeling is critical and helps understand the underlying biology of bone disorders. Initially, we have identified the ubiquitin-specific peptidase gene (Usp53) as a target of the parathyroid hormone in osteoblasts and a regulator of mesenchymal stem cell differentiation. Mutations in USP53 have been linked to a constellation of developmental pathologies. However, the role of Usp53 in bone has never been visited. Here we show that Usp53 null mice have a low bone mass phenotype in vivo. Usp53 null mice exhibit a pronounced decrease in trabecular bone indices including trabecular bone volume (36%) and trabecular number (26%) along with an increase in trabecular separation (13%). Cortical bone parameters are also impacted, showing a reduction in cortical bone volume (12%) and cortical bone thickness (15%). As a result, the strength and mechanical bone properties of Usp53 null mice have been compromised. At the cellular level, the ablation of Usp53 perturbs bone remodeling, augments osteoblast-dependent osteoclastogenesis, and increases osteoclast numbers. Bone marrow adipose tissue volume increased significantly with age in Usp53-deficient mice. Usp53 null mice displayed increased serum receptor activator of NF-κB ligand (RANKL) levels, and Usp53-deficient osteoblasts and bone marrow adipocytes have increased expression of Rankl. Mechanistically, USP53 regulates Rankl expression by enhancing the interaction between VDR and SMAD3. This is the first report describing the function of Usp53 during skeletal development. Our results put Usp53 in display as a novel regulator of osteoblast-osteoclast coupling and open the door for investigating the involvement of USP53 in pathologies. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Hadla Hariri
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada.,Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| | - Orhun Kose
- McGill Otolaryngology Sciences Laboratory, McGill University Health Centre-Research Institute, Montreal, Canada
| | - Aren Bezdjian
- McGill Otolaryngology Sciences Laboratory, McGill University Health Centre-Research Institute, Montreal, Canada
| | - Sam J Daniel
- McGill Otolaryngology Sciences Laboratory, McGill University Health Centre-Research Institute, Montreal, Canada.,Department of Otolaryngology-Head and Neck Surgery, McGill University, Montreal, Canada.,Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - René St-Arnaud
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada.,Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada.,Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada.,Department of Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
| |
Collapse
|
42
|
Panda DK, Bai X, Zhang Y, Stylianesis NA, Koromilas AE, Lipman ML, Karaplis AC. SCF-SKP2 E3 ubiquitin ligase links mTORC1/ER stress/ISR with YAP activation in murine renal cystogenesis. J Clin Invest 2022; 132:153943. [PMID: 36326820 PMCID: PMC9754004 DOI: 10.1172/jci153943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/21/2022] [Indexed: 11/06/2022] Open
Abstract
The Hippo pathway nuclear effector Yes-associated protein (YAP) potentiates the progression of polycystic kidney disease (PKD) arising from ciliopathies. The mechanisms underlying the increase in YAP expression and transcriptional activity in PKD remain obscure. We observed that in kidneys from mice with juvenile cystic kidney (jck) ciliopathy, the aberrant hyperactivity of mechanistic target of rapamycin complex 1 (mTORC1), driven by ERK1/2 and PI3K/AKT cascades, induced ER proteotoxic stress. To reduce this stress by reprogramming translation, the protein kinase R-like ER kinase-eukaryotic initiation factor 2α (PERK/eIF2α) arm of the integrated stress response (ISR) was activated. PERK-mediated phosphorylation of eIF2α drove the selective translation of activating transcription factor 4 (ATF4), potentiating YAP expression. In parallel, YAP underwent K63-linked polyubiquitination by SCF S-phase kinase-associated protein 2 (SKP2) E3 ubiquitin ligase, a Hippo-independent, nonproteolytic ubiquitination that enhances YAP nuclear trafficking and transcriptional activity in cancer cells. Defective ISR cellular adaptation to ER stress in eIF2α phosphorylation-deficient jck mice further augmented YAP-mediated transcriptional activity and renal cyst growth. Conversely, pharmacological tuning down of ER stress/ISR activity and SKP2 expression in jck mice by administration of tauroursodeoxycholic acid (TUDCA) or tolvaptan impeded these processes. Restoring ER homeostasis and/or interfering with the SKP2-YAP interaction represent potential therapeutic avenues for stemming the progression of renal cystogenesis.
Collapse
Affiliation(s)
- Dibyendu K. Panda
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital,,Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Xiuying Bai
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Yan Zhang
- Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | | | - Antonis E. Koromilas
- Gerald Bronfman Department of Oncology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Mark L. Lipman
- Division of Nephrology, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| | - Andrew C. Karaplis
- Division of Endocrinology and Metabolism, Department of Medicine, and Lady Davis Institute for Medical Research, Jewish General Hospital
| |
Collapse
|
43
|
Identification of purine biosynthesis as an NADH-sensing pathway to mediate energy stress. Nat Commun 2022; 13:7031. [PMID: 36396642 PMCID: PMC9672040 DOI: 10.1038/s41467-022-34850-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
An enhanced NADH/NAD+ ratio, termed reductive stress, is associated with many diseases. However, whether a downstream sensing pathway exists to mediate pathogenic outcomes remains unclear. Here, we generate a soluble pyridine nucleotide transhydrogenase from Escherichia coli (EcSTH), which can elevate the NADH/NAD+ ratio and meantime reduce the NADPH/NADP+ ratio. Additionally, we fuse EcSTH with previously described LbNOX (a water-forming NADH oxidase from Lactobacillus brevis) to resume the NADH/NAD+ ratio. With these tools and by using genome-wide CRISPR/Cas9 library screens and metabolic profiling in mammalian cells, we find that accumulated NADH deregulates PRPS2 (Ribose-phosphate pyrophosphokinase 2)-mediated downstream purine biosynthesis to provoke massive energy consumption, and therefore, the induction of energy stress. Blocking purine biosynthesis prevents NADH accumulation-associated cell death in vitro and tissue injury in vivo. These results underscore the pathophysiological role of deregulated purine biosynthesis in NADH accumulation-associated disorders and demonstrate the utility of EcSTH in manipulating NADH/NAD+ and NADPH/NADP+.
Collapse
|
44
|
Hannan KM, Soo P, Wong MS, Lee JK, Hein N, Poh P, Wysoke KD, Williams TD, Montellese C, Smith LK, Al-Obaidi SJ, Núñez-Villacís L, Pavy M, He JS, Parsons KM, Loring KE, Morrison T, Diesch J, Burgio G, Ferreira R, Feng ZP, Gould CM, Madhamshettiwar PB, Flygare J, Gonda TJ, Simpson KJ, Kutay U, Pearson RB, Engel C, Watkins NJ, Hannan RD, George AJ. Nuclear stabilization of p53 requires a functional nucleolar surveillance pathway. Cell Rep 2022; 41:111571. [DOI: 10.1016/j.celrep.2022.111571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/06/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
|
45
|
Luo X, Song S, Qi L, Tien CL, Li H, Xu W, Mathuram TL, Burris T, Zhao Y, Sun Z, Zhang L. REV-ERB is essential in cardiac fibroblasts homeostasis. Front Pharmacol 2022; 13:899628. [PMID: 36386186 PMCID: PMC9662302 DOI: 10.3389/fphar.2022.899628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/10/2022] [Indexed: 01/28/2023] Open
Abstract
REV-ERB agonists have shown antifibrotic effects in the heart and other organs. The function of REV-ERB in the cardiac fibroblasts remains unstudied. Here, we characterize the functional difference of REV-ERB in mouse embryonic fibroblasts and cardiac fibroblasts using genetic deletion of REV-ERBα and ß in vitro. We show that REV-ERB α/β double deleted cardiac fibroblasts have reduced viability and proliferation, but increased migration and myofibroblasts activation. Thus, REV-ERB α/β has essential cell-autonomous role in cardiac fibroblasts in maintaining them in a healthy, quiescent state. We also show that existing REV-ERB agonist SR9009 strongly suppresses cardiac fibroblasts activation but in a REV-ERB-independent manner highlighting the need to develop novel REV-ERB agonists for treating cardiac fibrosis.
Collapse
Affiliation(s)
- Xiaokang Luo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Shiyang Song
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, United States
| | - Lei Qi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Chih-Liang Tien
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Hui Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Theodore Lemuel Mathuram
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Thomas Burris
- Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Yuanbiao Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Zheng Sun
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
46
|
Mapping autophagosome contents identifies interleukin-7 receptor-α as a key cargo modulating CD4+ T cell proliferation. Nat Commun 2022; 13:5174. [PMID: 36055998 PMCID: PMC9440129 DOI: 10.1038/s41467-022-32718-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
CD4+ T cells are pivotal cells playing roles in the orchestration of humoral and cytotoxic immune responses. It is known that CD4+ T cell proliferation relies on autophagy, but identification of the autophagosomal cargo involved is missing. Here we create a transgenic mouse model, to enable direct mapping of the proteinaceous content of autophagosomes in primary cells by LC3 proximity labelling. Interleukin-7 receptor-α, a cytokine receptor mostly found in naïve and memory T cells, is reproducibly detected in autophagosomes of activated CD4+ T cells. Consistently, CD4+ T cells lacking autophagy show increased interleukin-7 receptor-α surface expression, while no defect in internalisation is observed. Mechanistically, excessive surface interleukin-7 receptor-α sequestrates the common gamma chain, impairing the interleukin-2 receptor assembly and downstream signalling crucial for T cell proliferation. This study shows that key autophagy substrates can be reliably identified in this mouse model and help mechanistically unravel autophagy's contribution to healthy physiology and disease.
Collapse
|
47
|
Yu T, Slone J, Liu W, Barnes R, Opresko PL, Wark L, Mai S, Horvath S, Huang T. Premature aging is associated with higher levels of 8-oxoguanine and increased DNA damage in the Polg mutator mouse. Aging Cell 2022; 21:e13669. [PMID: 35993394 PMCID: PMC9470903 DOI: 10.1111/acel.13669] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 01/24/2023] Open
Abstract
Mitochondrial dysfunction plays an important role in the aging process. However, the mechanism by which this dysfunction causes aging is not fully understood. The accumulation of mutations in the mitochondrial genome (or "mtDNA") has been proposed as a contributor. One compelling piece of evidence in support of this hypothesis comes from the PolgD257A/D257A mutator mouse (Polgmut/mut ). These mice express an error-prone mitochondrial DNA polymerase that results in the accumulation of mtDNA mutations, accelerated aging, and premature death. In this paper, we have used the Polgmut/mut model to investigate whether the age-related biological effects observed in these mice are triggered by oxidative damage to the DNA that compromises the integrity of the genome. Our results show that mutator mouse has significantly higher levels of 8-oxoguanine (8-oxoGua) that are correlated with increased nuclear DNA (nDNA) strand breakage and oxidative nDNA damage, shorter average telomere length, and reduced mtDNA integrity. Based on these results, we propose a model whereby the increased level of reactive oxygen species (ROS) associated with the accumulation of mtDNA mutations in Polgmut/mut mice results in higher levels of 8-oxoGua, which in turn lead to compromised DNA integrity and accelerated aging via increased DNA fragmentation and telomere shortening. These results suggest that mitochondrial play a central role in aging and may guide future research to develop potential therapeutics for mitigating aging process.
Collapse
Affiliation(s)
- Tenghui Yu
- Department of PediatricsUniversity at BuffaloBuffaloNew YorkUSA,Human Aging Research Institute, School of Life ScienceNanchang UniversityNanchangChina,Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Jesse Slone
- Department of PediatricsUniversity at BuffaloBuffaloNew YorkUSA,Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Wensheng Liu
- Department of PediatricsUniversity at BuffaloBuffaloNew YorkUSA
| | - Ryan Barnes
- Department of Environmental and Occupational HealthUniversity of Pittsburgh Graduate School of Public Health, and UPMC Hillman Cancer CenterPittsburghPennsylvaniaUSA
| | - Patricia L. Opresko
- Department of Environmental and Occupational HealthUniversity of Pittsburgh Graduate School of Public Health, and UPMC Hillman Cancer CenterPittsburghPennsylvaniaUSA
| | - Landon Wark
- CancerCare Manitoba Research Institute, The Genomic Center for Cancer Research & DiagnosisUniversity of ManitobaWinnipegManitobaCanada
| | - Sabine Mai
- CancerCare Manitoba Research Institute, The Genomic Center for Cancer Research & DiagnosisUniversity of ManitobaWinnipegManitobaCanada
| | - Steve Horvath
- Human Genetics, David Geffen School of MedicineUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Taosheng Huang
- Department of PediatricsUniversity at BuffaloBuffaloNew YorkUSA,Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| |
Collapse
|
48
|
Noncanonical PDK4 action alters mitochondrial dynamics to affect the cellular respiratory status. Proc Natl Acad Sci U S A 2022; 119:e2120157119. [PMID: 35969774 PMCID: PMC9407676 DOI: 10.1073/pnas.2120157119] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dynamic regulation of mitochondrial morphology provides cells with the flexibility required to adapt and respond to electron transport chain (ETC) toxins and mitochondrial DNA-linked disease mutations, yet the mechanisms underpinning the regulation of mitochondrial dynamics machinery by these stimuli is poorly understood. Here, we show that pyruvate dehydrogenase kinase 4 (PDK4) is genetically required for cells to undergo rapid mitochondrial fragmentation when challenged with ETC toxins. Moreover, PDK4 overexpression was sufficient to promote mitochondrial fission even in the absence of mitochondrial stress. Importantly, we observed that the PDK4-mediated regulation of mitochondrial fission was independent of its canonical function, i.e., inhibitory phosphorylation of the pyruvate dehydrogenase complex (PDC). Phosphoproteomic screen for PDK4 substrates, followed by nonphosphorylatable and phosphomimetic mutations of the PDK4 site revealed cytoplasmic GTPase, Septin 2 (SEPT2), as the key effector molecule that acts as a receptor for DRP1 in the outer mitochondrial membrane to promote mitochondrial fission. Conversely, inhibition of the PDK4-SEPT2 axis could restore the balance in mitochondrial dynamics and reinvigorates cellular respiration in mitochondrial fusion factor, mitofusin 2-deficient cells. Furthermore, PDK4-mediated mitochondrial reshaping limits mitochondrial bioenergetics and supports cancer cell growth. Our results identify the PDK4-SEPT2-DRP1 axis as a regulator of mitochondrial function at the interface between cellular bioenergetics and mitochondrial dynamics.
Collapse
|
49
|
Walter F, D’Orsi B, Jagannathan A, Dussmann H, Prehn JHM. BOK controls ER proteostasis and physiological ER stress responses in neurons. Front Cell Dev Biol 2022; 10:915065. [PMID: 36060797 PMCID: PMC9434404 DOI: 10.3389/fcell.2022.915065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The Bcl-2 family proteins BAK and BAX control the crucial step of pore formation in the mitochondrial outer membrane during intrinsic apoptosis. Bcl-2-related ovarian killer (BOK) is a Bcl-2 family protein with a high sequence similarity to BAK and BAX. However, intrinsic apoptosis can proceed in the absence of BOK. Unlike BAK and BAX, BOK is primarily located on the endoplasmic reticulum (ER) and Golgi membranes, suggesting a role for BOK in regulating ER homeostasis. In this study, we report that BOK is required for a full ER stress response. Employing previously characterized fluorescent protein-based ER stress reporter cell systems, we show that BOK-deficient cells have an attenuated response to ER stress in all three signaling branches of the unfolded protein response. Fluo-4-based confocal Ca2+ imaging revealed that disruption of ER proteostasis in BOK-deficient cells was not linked to altered ER Ca2+ levels. Fluorescence recovery after photobleaching (FRAP) experiments using GRP78/BiP-eGFP demonstrated that GRP78 motility was significantly lower in BOK-deficient cells. This implied that less intraluminal GRP78 was freely available and more of the ER chaperone bound to unfolded proteins. Collectively, these experiments suggest a new role for BOK in the protection of ER proteostasis and cellular responses to ER stress.
Collapse
Affiliation(s)
- Franziska Walter
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Beatrice D’Orsi
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- Institute of Neuroscience, Italian National Research Council, Pisa, Italy
| | - Anagha Jagannathan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Jochen H. M. Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- SFI FutureNeuro Research Centre, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
- *Correspondence: Jochen H. M. Prehn,
| |
Collapse
|
50
|
A novel Gboxin analog induces OXPHOS inhibition and mitochondrial dysfunction-mediated apoptosis in diffuse large B-cell lymphoma. Bioorg Chem 2022; 127:106019. [PMID: 35849895 DOI: 10.1016/j.bioorg.2022.106019] [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: 04/20/2022] [Revised: 06/07/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive B-cell non-Hodgkin's lymphoma. Currently, moderate efficacy and limitations of approved drugs still exist, and it is necessary to develop newer and more effective drugs. Gboxin is a promising inhibitor of OXPHOS, which specifically inhibits the growth of many kinds of cancer cell lines. In the present study, 21 Gboxin analogs incorporating amide and ester moieties were designed and synthesized. Preliminary screening results show that 5d also has specific selectivity for cancer cells, particularly on the DLBCL cells, which is weaker than that of Gboxin but still good. Thus, the effect and underlying mechanism of 5d on DLBCL cells were further studied. The results showed that 5d exhibits potent proliferation inhibition and cell cycle arrest effects, and its IC50 to DLBCL cells is below 1 µM. In addition, 5d induces apoptosis of DLBCL cells in a time- and dose-dependent manner, and this effect is stronger than that of Gboxin and VP16. Mechanistically, 5d plays its role mainly through the stimulation of metabolic stress in DLBCL cell lines, which induces OXPHOS inhibition, inflammation, DNA damage and mitochondrial dysfunction. These data suggest that 5d has potential as a candidate agent for DLBCL alternative drug development.
Collapse
|