1
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Aggravating mechanisms from COVID-19. Virol J 2024; 21:228. [PMID: 39334442 PMCID: PMC11430051 DOI: 10.1186/s12985-024-02506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces immune-mediated diseases. The pathophysiology of COVID-19 uses the following three mechanisms: (1) inflammasome activation mechanism; (2) cGAS-STING signaling mechanism; and (3) SAMHD1 tetramerization mechanism, which leads to IFN-I production. Interactions between the host and virus govern induction, resulting in multiorgan impacts. The NLRP3 with cGAS-STING constitutes the primary immune response. The expression of SARS-CoV-2 ORF3a, NSP6, NSP7, and NSP8 blocks innate immune activation and facilitates virus replication by targeting the RIG-I/MDA5, TRIF, and cGAS-STING signaling. SAMHD1 has a target motif for CDK1 to protect virion assembly, threonine 592 to modulate a catalytically active tetramer, and antiviral IFN responses to block retroviral infection. Plastic and allosteric nucleic acid binding of SAMHD1 modulates the antiretroviral activity of SAMHD1. Therefore, inflammasome activation, cGAS-STING signaling, and SAMHD1 tetramerization explain acute kidney injury, hepatic, cardiac, neurological, and gastrointestinal injury of COVID-19. It might be necessary to effectively block the pathological courses of diverse diseases.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Geriatrics, Gyeonggi Medical Center Pocheon Hospital, 1648 Pocheon-ro Sin-eup-dong, Pocheon-si, Gyeonggi-do, 11142, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408 VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
2
|
He Y, Zhou L, Wang M, Zhong Z, Chen H, Lian C, Zhang H, Wang H, Cao L, Li C. Integrated transcriptomic and metabolomic approaches reveal molecular response and potential biomarkers of the deep-sea mussel Gigantidas platifrons to copper exposure. JOURNAL OF HAZARDOUS MATERIALS 2024; 473:134612. [PMID: 38761766 DOI: 10.1016/j.jhazmat.2024.134612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/27/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Metal pollution caused by deep-sea mining activities has potential detrimental effects on deep-sea ecosystems. However, our knowledge of how deep-sea organisms respond to this pollution is limited, given the challenges of remoteness and technology. To address this, we conducted a toxicity experiment by using deep-sea mussel Gigantidas platifrons as model animals and exposing them to different copper (Cu) concentrations (50 and 500 μg/L) for 7 days. Transcriptomics and LC-MS-based metabolomics methods were employed to characterize the profiles of transcription and metabolism in deep-sea mussels exposed to Cu. Transcriptomic results suggested that Cu toxicity significantly affected the immune response, apoptosis, and signaling processes in G. platifrons. Metabolomic results demonstrated that Cu exposure disrupted its carbohydrate metabolism, anaerobic metabolism and amino acid metabolism. By integrating both sets of results, transcriptomic and metabolomic, we find that Cu exposure significantly disrupts the metabolic pathway of protein digestion and absorption in G. platifrons. Furthermore, several key genes (e.g., heat shock protein 70 and baculoviral IAP repeat-containing protein 2/3) and metabolites (e.g., alanine and succinate) were identified as potential molecular biomarkers for deep-sea mussel's responses to Cu toxicity. This study contributes novel insight for assessing the potential effects of deep-sea mining activities on deep-sea organisms.
Collapse
Affiliation(s)
- Yameng He
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Li Zhou
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China.
| | - Minxiao Wang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Zhaoshan Zhong
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Hao Chen
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Chao Lian
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Huan Zhang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Hao Wang
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Lei Cao
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Chaolun Li
- Center of Deep Sea Research, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; University of Chinese Academy of Sciences, Beijing 10049, China; Laoshan Laboratory, Qingdao 266237, China.
| |
Collapse
|
3
|
Yang J, Cui L, Zhang Y, Ling Z, Zhang Z, Huang Y, Ma J, Xiao S, Yang B, Huang L. Unravelling the genetic basis for skeletal muscle mitochondrial DNA copy number variations in pigs. SCIENCE CHINA. LIFE SCIENCES 2024; 67:211-214. [PMID: 37906412 DOI: 10.1007/s11427-022-2397-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Indexed: 11/02/2023]
Affiliation(s)
- Jiawen Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Leilei Cui
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
- School of Life Sciences, Nanchang University, Nanchang, 330047, China
| | - Yifeng Zhang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ziqi Ling
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhou Zhang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Yizhong Huang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Junwu Ma
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shijun Xiao
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Bin Yang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Lusheng Huang
- National Key Laboratory for Swine Genetic Improvement and Germplasm Innovation, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
4
|
Silva MM, Campos TA, Cavalcanti IMF, Oliveira IS, Pérez CD, Silva RADA, Wanderley MSO, Santos NPS. Proteomic characterization and biological activities of the mucus produced by the zoanthid Palythoa caribaeorum (Duchassaing & Michelotti, 1860). AN ACAD BRAS CIENC 2023; 95:e20200325. [PMID: 38055606 DOI: 10.1590/0001-3765202320200325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/22/2020] [Indexed: 12/08/2023] Open
Abstract
Mucus, produced by Palythoa caribaeorum has been popularly reported due to healing, anti-inflammatory, and analgesic effects. However, biochemical and pharmacological properties of this mucus remains unexplored. Therefore, the present study aimed to study its proteome profile by 2DE electrophoresis and MALDI-TOF. Furthermore, it was evaluated the cytotoxic, antibacterial, and antioxidant activities of the mucus and from its protein extract (PE). Proteomics study identified14 proteins including proteins involved in the process of tissue regeneration and death of tumor cells. The PE exhibited cell viability below 50% in the MCF-7 and S-180 strains. It showed IC50 of 6.9 μg/mL for the J774 lineage, and also, favored the cellular growth of fibroblasts. Furthermore, PE revealed activity against Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus, and Staphylococcus epidermidis (MIC of 250 μg/mL). These findings revealed the mucus produced by Palythoa caribaeorum with biological activities, offering alternative therapies for the treatment of cancer and as a potential antibacterial agent.
Collapse
Affiliation(s)
- Marllyn M Silva
- Universidade Federal de Pernambuco, Centro Acadêmico de Vitória, Núcleo de Biologia, Rua Alto do Reservatório, s/n, Bela Vista, 55608-680 Vitória de Santo Antão, PE, Brazil
| | - Thiers A Campos
- Centro Tecnológico do Nordeste, Av. Prof. Luís Freire, 1, Cidade Universitária, 50740-545 Recife, PE, Brazil
| | - Isabella M F Cavalcanti
- Universidade Federal de Pernambuco, Centro Acadêmico de Vitória, Núcleo de Biologia, Rua Alto do Reservatório, s/n, Bela Vista, 55608-680 Vitória de Santo Antão, PE, Brazil
- Universidade Federal de Pernambuco, Instituto Keizo-Asami (iLIKA), Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Idjane S Oliveira
- Universidade Federal de Pernambuco, Centro Acadêmico de Vitória, Núcleo de Biologia, Rua Alto do Reservatório, s/n, Bela Vista, 55608-680 Vitória de Santo Antão, PE, Brazil
| | - Carlos Daniel Pérez
- Universidade Federal de Pernambuco, Centro Acadêmico de Vitória, Núcleo de Biologia, Rua Alto do Reservatório, s/n, Bela Vista, 55608-680 Vitória de Santo Antão, PE, Brazil
| | - Roberto Afonso DA Silva
- Universidade Federal de Pernambuco, Instituto Keizo-Asami (iLIKA), Av. Prof. Moraes Rego, s/n, Cidade Universitária, 50670-901 Recife, PE, Brazil
| | - Marcela S O Wanderley
- Universidade de Pernambuco, Campus Santo Amaro, Instituto de Ciências Biológicas, Arnóbio Marques, 310, Santo Amaro, 50100-130 Recife, PE, Brazil
| | - Noemia P S Santos
- Universidade Federal de Pernambuco, Centro Acadêmico de Vitória, Núcleo de Biologia, Rua Alto do Reservatório, s/n, Bela Vista, 55608-680 Vitória de Santo Antão, PE, Brazil
| |
Collapse
|
5
|
Xu B, Sui Q, Hu H, Hu X, Zhou X, Qian C, Li N. SAMHD1 Attenuates Acute Inflammation by Maintaining Mitochondrial Function in Macrophages via Interaction with VDAC1. Int J Mol Sci 2023; 24:7888. [PMID: 37175593 PMCID: PMC10177872 DOI: 10.3390/ijms24097888] [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: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Over-activation of Toll-like receptor 4 (TLR4) is the key mechanism in Gram-negative bacterial infection-induced sepsis. SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1 (SAMHD1) inhibits multiple viruses, but whether it plays a role during bacterial invasion remains unelucidated. Monocyte-macrophage specific Samhd1 knockout (Samhd1-/-) mice and Samhd1-/- macrophage cell line RAW264.7 were constructed and used as research models to evaluate the role of SAMHD1 in TLR4-activated inflammation. In vivo, LPS-challenged Samhd1-/- mice showed higher serum inflammatory factors, accompanied with more severe inflammation infiltration and lower survival rate. In vitro, Samhd1-/- peritoneal macrophages had more activated TLR4 pathway upon LPS-stimulation, accompanied with mitochondrial depolarization and dysfunction and a higher tendency to be M1-polarized. These results could be rescued by overexpressing full-length wild-type SAMHD1 or its phospho-mimetic T634D mutant into Samhd1-/- RAW264.7 cells, whereas the mutants, dNTP hydrolase-function-deprived H238A and phospho-ablative T634A, did not exert the same effect. Lastly, co-IP and immunofluorescence assays confirmed that SAMHD1 interacted with an outer mitochondrial membrane-localized protein, voltage-dependent anion channel-1 (VDAC1). SAMHD1 inhibits TLR4-induced acute inflammation and M1 polarization of macrophages by interacting with VDAC1 and maintaining mitochondria function, which outlines a novel regulatory mechanism of TLR signaling upon LPS stimulation.
Collapse
Affiliation(s)
- Bowen Xu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Qianyi Sui
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Han Hu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiangjia Hu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Xuchang Zhou
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Cheng Qian
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Nan Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
6
|
D'Aronco G, Ferraro P, Sassano V, Dagostino C, Biancotto M, Palumbo E, Presot E, Russo A, Bianchi V, Rampazzo C. SAMHD1 restricts the deoxyguanosine triphosphate pool contributing to telomere stability in telomerase-positive cells. FASEB J 2023; 37:e22883. [PMID: 36934410 PMCID: PMC11977530 DOI: 10.1096/fj.202300122r] [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: 01/23/2023] [Revised: 02/26/2023] [Accepted: 03/07/2023] [Indexed: 03/20/2023]
Abstract
SAMHD1 (Sterile alpha motif and histidine/aspartic acid domain-containing protein 1) is a dNTP triphosphohydrolase crucial in the maintenance of balanced cellular dNTP pools, which support genome integrity. In SAMHD1 deficient fibroblasts isolated from Aicardi-Goutières Syndrome (AGS) patients, all four DNA precursors are increased and markedly imbalanced with the largest effect on dGTP, a key player in the modulation of telomerase processivity. Here, we present data showing that SAMHD1, by restricting the dGTP pool, contributes to telomere maintenance in hTERT-immortalized human fibroblasts from AGS patients as well as in telomerase positive cancer cell lines. Only in cells expressing telomerase, the lack of SAMHD1 causes excessive lengthening of telomeres and telomere fragility, whereas primary fibroblasts lacking both SAMHD1 and telomerase enter normally into senescence. Telomere lengthening observed in SAMHD1 deficient but telomerase proficient cells is a gradual process, in accordance with the intrinsic property of telomerase of adding only a few tens of nucleotides for each cycle. Therefore, only a prolonged exposure to high dGTP content causes telomere over-elongation. hTERT-immortalized AGS fibroblasts display also high fragility of chromosome ends, a marker of telomere replication stress. These results not only demonstrate the functional importance of dGTP cellular level but also reveal the critical role played by SAMHD1 in restraining telomerase processivity and safeguarding telomere stability.
Collapse
Affiliation(s)
| | - Paola Ferraro
- Department of BiologyUniversity of PadovaPadovaItaly
| | | | | | | | - Elisa Palumbo
- Department of Molecular MedicineUniversity of PadovaPadovaItaly
| | - Ettore Presot
- Department of BiologyUniversity of PadovaPadovaItaly
| | - Antonella Russo
- Department of Molecular MedicineUniversity of PadovaPadovaItaly
| | - Vera Bianchi
- Department of BiologyUniversity of PadovaPadovaItaly
| | | |
Collapse
|
7
|
Lu C, Wang Q, Jiang Y, Zhang M, Meng X, Li Y, Liu B, Yin Z, Liu H, Peng C, Li F, Yue Y, Hao M, Sui Y, Wang L, Cheng G, Liu J, Chu Z, Zhu C, Dong H, Ding X. Discovery of a novel nucleoside immune signaling molecule 2'-deoxyguanosine in microbes and plants. J Adv Res 2023; 46:1-15. [PMID: 35811061 PMCID: PMC10105077 DOI: 10.1016/j.jare.2022.06.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/16/2022] [Accepted: 06/27/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Beneficial microorganisms play essential roles in plant growth and induced systemic resistance (ISR) by releasing signaling molecules. Our previous study obtained the crude extract from beneficial endophyte Paecilomyces variotii, termed ZNC (ZhiNengCong), which significantly enhanced plant resistance to pathogen even at 100 ng/ml. However, the immunoreactive components of ZNC remain unclear. Here, we further identified one of the immunoreactive components of ZNC is a nucleoside 2'-deoxyguanosine (2-dG). OBJECTIVES This paper intends to reveal the molecular mechanism of microbial-derived 2'-deoxyguanosine (2-dG) in activating plant immunity, and the role of plant-derived 2-dG in plant immunity. METHODS The components of ZNC were separated using a high-performance liquid chromatography (HPLC), and 2-dG is identified using a HPLC-mass spectrometry system (LC-MS). Transcriptome analysis and genetic experiments were used to reveal the immune signaling pathway dependent on 2-dG activation of plant immunity. RESULTS This study identified 2'-deoxyguanosine (2-dG) as one of the immunoreactive components from ZNC. And 2-dG significantly enhanced plant pathogen resistance even at 10 ng/ml (37.42 nM). Furthermore, 2-dG-induced resistance depends on NPR1, pattern-recognition receptors/coreceptors, ATP receptor P2K1 (DORN1), ethylene signaling but not salicylic acid accumulation. In addition, we identified Arabidopsis VENOSA4 (VEN4) was involved in 2-dG biosynthesis and could convert dGTP to 2-dG, and vne4 mutant plants were more susceptible to pathogens. CONCLUSION In summary, microbial-derived 2-dG may act as a novel immune signaling molecule involved in plant-microorganism interactions, and VEN4 is 2-dG biosynthesis gene and plays a key role in plant immunity.
Collapse
Affiliation(s)
- Chongchong Lu
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Qingbin Wang
- Shandong Pengbo Biotechnology Co., LTD, Taian 271018, China; National Engineering Laboratory for Efficient Utilization of Soil and Fertilizer Resources, College of Resources and Environment, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yanke Jiang
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Min Zhang
- National Engineering Laboratory for Efficient Utilization of Soil and Fertilizer Resources, College of Resources and Environment, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xuanlin Meng
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yang Li
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Baoyou Liu
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Ziyi Yin
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Haifeng Liu
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chune Peng
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Fuchuan Li
- National Glycoengineering Research Center and Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, 72 Binhai Rd, Qingdao 266200, China
| | - Yingzhe Yue
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Mingxia Hao
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yurong Sui
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Lulu Wang
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Guodong Cheng
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhaohui Chu
- State Key Laboratory of Hybrid Rice, Hubei Hongshan Laboratory, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Changxiang Zhu
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Hansong Dong
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Xinhua Ding
- State Key Laboratory of Crop Biology, Shandong Provincial Key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
8
|
Pillalamarri V, Shi W, Say C, Yang S, Lane J, Guallar E, Pankratz N, Arking DE. Whole-exome sequencing in 415,422 individuals identifies rare variants associated with mitochondrial DNA copy number. HGG ADVANCES 2023; 4:100147. [PMID: 36311265 PMCID: PMC9615038 DOI: 10.1016/j.xhgg.2022.100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/23/2022] [Indexed: 10/14/2022] Open
Abstract
Inter-individual variation in the number of copies of the mitochondrial genome, called mitochondrial DNA copy number (mtDNA-CN), reflects mitochondrial function and has been associated with various aging-related diseases. We examined 415,422 exomes of self-reported White ancestry individuals from the UK Biobank and tested the impact of rare variants, at the level of single variants and through aggregate variant-set tests, on mtDNA-CN. A survey across nine variant sets tested enrichment of putatively causal variants and identified 14 genes at experiment-wide significance and three genes at marginal significance. These included associations at known mtDNA depletion syndrome genes (mtDNA helicase TWNK, p = 1.1 × 10-30; mitochondrial transcription factor TFAM, p = 4.3 × 10-15; mtDNA maintenance exonuclease MGME1, p = 2.0 × 10-6) and the V617F dominant gain-of-function mutation in the tyrosine kinase JAK2 (p = 2.7 × 10-17), associated with myeloproliferative disease. Novel genes included the ATP-dependent protease CLPX (p = 8.4 × 10-9), involved in mitochondrial proteome quality, and the mitochondrial adenylate kinase AK2 (p = 4.7 × 10-8), involved in hematopoiesis. The most significant association was a missense variant in SAMHD1 (p = 4.2 × 10-28), found on a rare, 1.2-Mb shared ancestral haplotype on chromosome 20. SAMHD1 encodes a cytoplasmic host restriction factor involved in viral defense response and the mitochondrial nucleotide salvage pathway, and is associated with Aicardi-Goutières syndrome 5, a childhood encephalopathy and chronic inflammatory response disorder. Rare variants were enriched in Mendelian mtDNA depletion syndrome loci, and these variants implicated core processes in mtDNA replication, nucleoid structure formation, and maintenance. These data indicate that strong-effect mutations from the nuclear genome contribute to the genetic architecture of mtDNA-CN.
Collapse
Affiliation(s)
- Vamsee Pillalamarri
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Predoctoral Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Maryland Genetics Epidemiology and Medicine Training Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Wen Shi
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Conrad Say
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephanie Yang
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Vertex Pharmaceuticals, Inc., Boston, MA 02210, USA
| | - John Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Eliseo Guallar
- Departments of Epidemiology and Medicine and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Dan E. Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
9
|
Levi J, Song H. The other immuno-PET: Metabolic tracers in evaluation of immune responses to immune checkpoint inhibitor therapy for solid tumors. Front Immunol 2023; 13:1113924. [PMID: 36700226 PMCID: PMC9868703 DOI: 10.3389/fimmu.2022.1113924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Unique patterns of response to immune checkpoint inhibitor therapy, discernable in the earliest clinical trials, demanded a reconsideration of the standard methods of radiological treatment assessment. Immunomonitoring, that characterizes immune responses, offers several significant advantages over the tumor-centric approach currently used in the clinical practice: 1) better understanding of the drugs' mechanism of action and treatment resistance, 2) earlier assessment of response to therapy, 3) patient/therapy selection, 4) evaluation of toxicity and 5) more accurate end-point in clinical trials. PET imaging in combination with the right agent offers non-invasive tracking of immune processes on a whole-body level and thus represents a method uniquely well-suited for immunomonitoring. Small molecule metabolic tracers, largely neglected in the immuno-PET discourse, offer a way to monitor immune responses by assessing cellular metabolism known to be intricately linked with immune cell function. In this review, we highlight the use of small molecule metabolic tracers in imaging immune responses, provide a view of their value in the clinic and discuss the importance of image analysis in the context of tracking a moving target.
Collapse
Affiliation(s)
- Jelena Levi
- CellSight Technologies Incorporated, San Francisco, CA, United States,*Correspondence: Jelena Levi,
| | - Hong Song
- Department of Radiology, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
10
|
Biodistribution of a Mitochondrial Metabolic Tracer, [ 18F]F-AraG, in Healthy Volunteers. Mol Imaging 2022; 2022:3667417. [PMID: 36072652 PMCID: PMC9400547 DOI: 10.1155/2022/3667417] [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: 06/03/2022] [Accepted: 07/25/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose [18F]F-AraG is a radiolabeled nucleoside analog that shows relative specificity for activated T cells. The aim of this study was to investigate the biodistribution of [18F]F-AraG in healthy volunteers and assess the preliminary safety and radiation dosimetry. Methods Six healthy subjects (three female and three male) between the ages of 24 and 60 participated in the study. Each subject received a bolus venous injection of [18F]F-AraG (dose range: 244.2-329.3 MBq) prior to four consecutive PET/MR whole-body scans. Blood samples were collected at regular intervals and vital signs monitored before and after tracer administration. Regions of interest were delineated for multiple organs, and the area under the time-activity curves was calculated for each organ and used to derive time-integrated activity coefficient (TIAC). TIACs were input for absorbed dose and effective dose calculations using OLINDA. Results PET/MR examination was well tolerated, and no adverse effects to the administration of [18F]F-AraG were noted by the study participants. The biodistribution was generally reflective of the expression and activity profiles of the enzymes involved in [18F]F-AraG's cellular accumulation, mitochondrial kinase dGK, and SAMHD1. The highest uptake was observed in the kidneys and liver, while the brain, lung, bone marrow, and muscle showed low tracer uptake. The estimated effective dose for [18F]F-AraG was 0.0162 mSv/MBq (0.0167 mSv/MBq for females and 0.0157 mSv/MBq for males). Conclusion Biodistribution of [18F]F-AraG in healthy volunteers was consistent with its association with mitochondrial metabolism. PET/MR [18F]F-AraG imaging was well tolerated, with a radiation dosimetry profile similar to other commonly used [18F]-labeled tracers. [18F]F-AraG's connection with mitochondrial biogenesis and favorable biodistribution characteristics make it an attractive tracer with a variety of potential applications.
Collapse
|
11
|
López-Gómez C, Cámara Y, Hirano M, Martí R. 232nd ENMC international workshop: Recommendations for treatment of mitochondrial DNA maintenance disorders. 16 - 18 June 2017, Heemskerk, The Netherlands. Neuromuscul Disord 2022; 32:609-620. [PMID: 35641351 DOI: 10.1016/j.nmd.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/24/2022]
Affiliation(s)
| | - Yolanda Cámara
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain; Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Michio Hirano
- Columbia University Irving Medical Center, New York, USA
| | - Ramon Martí
- Research Group on Neuromuscular and Mitochondrial Diseases, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain; Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
12
|
Wang H, He X, Zhang L, Dong H, Huang F, Xian J, Li M, Chen W, Lu X, Pathak KV, Huang W, Li Z, Zhang L, Nguyen LXT, Yang L, Feng L, Gordon DJ, Zhang J, Pirrotte P, Chen CW, Salhotra A, Kuo YH, Horne D, Marcucci G, Sykes DB, Tiziani S, Jin H, Wang X, Li L. Disruption of dNTP homeostasis by ribonucleotide reductase hyperactivation overcomes AML differentiation blockade. Blood 2022; 139:3752-3770. [PMID: 35439288 PMCID: PMC9247363 DOI: 10.1182/blood.2021015108] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/07/2022] [Indexed: 01/09/2023] Open
Abstract
Differentiation blockade is a hallmark of acute myeloid leukemia (AML). A strategy to overcome such a blockade is a promising approach against the disease. The lack of understanding of the underlying mechanisms hampers development of such strategies. Dysregulated ribonucleotide reductase (RNR) is considered a druggable target in proliferative cancers susceptible to deoxynucleoside triphosphate (dNTP) depletion. Herein, we report an unanticipated discovery that hyperactivating RNR enables differentiation and decreases leukemia cell growth. We integrate pharmacogenomics and metabolomics analyses to identify that pharmacologically (eg, nelarabine) or genetically upregulating RNR subunit M2 (RRM2) creates a dNTP pool imbalance and overcomes differentiation arrest. Moreover, R-loop-mediated DNA replication stress signaling is responsible for RRM2 activation by nelarabine treatment. Further aggravating dNTP imbalance by depleting the dNTP hydrolase SAM domain and HD domain-containing protein 1 (SAMHD1) enhances ablation of leukemia stem cells by RRM2 hyperactivation. Mechanistically, excessive activation of extracellular signal-regulated kinase (ERK) signaling downstream of the imbalance contributes to cellular outcomes of RNR hyperactivation. A CRISPR screen identifies a synthetic lethal interaction between loss of DUSP6, an ERK-negative regulator, and nelarabine treatment. These data demonstrate that dNTP homeostasis governs leukemia maintenance, and a combination of DUSP inhibition and nelarabine represents a therapeutic strategy.
Collapse
Affiliation(s)
- Hanying Wang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Medical Oncology and
| | - Xin He
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lei Zhang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Haojie Dong
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Feiteng Huang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Jie Xian
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Min Li
- Department of Information Sciences, Beckman Research Institute and
| | - Wei Chen
- Integrative Genomics Core, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Xiyuan Lu
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
| | - Khyatiben V Pathak
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
| | - Wenfeng Huang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Zheng Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lianjun Zhang
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Le Xuan Truong Nguyen
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lu Yang
- Department of Systems Biology, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - Lifeng Feng
- Laboratory of Cancer Biology, Provincial Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - David J Gordon
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, University of Iowa, Iowa City, IA
| | - Jing Zhang
- McArdle Laboratory for Cancer Research and Wisconsin Blood Cancer Research Institute, University of Wisconsin-Madison, Madison, WI
| | - Patrick Pirrotte
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
- Cancer & Cell Biology Division, The Translational Genomics Research Institute, Phoenix, AZ
| | - Chun-Wei Chen
- Department of Systems Biology, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | | | - Ya-Huei Kuo
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte, CA
| | - Guido Marcucci
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
- Department of Hematology and Hematopoietic Cell Transplantation and
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA; and
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX
- Department of Pediatrics and
- Department of Oncology, Dell Medical School, LiveSTRONG Cancer Institutes, The University of Texas at Austin, Austin, TX
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Provincial Key Laboratory of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | | | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA
| |
Collapse
|
13
|
Fang H, Xie A, Du M, Li X, Yang K, Fu Y, Yuan X, Fan R, Yu W, Zhou Z, Sang T, Nie K, Li J, Zhao Q, Chen Z, Yang Y, Hong C, Lyu J. SERAC1 is a component of the mitochondrial serine transporter complex required for the maintenance of mitochondrial DNA. Sci Transl Med 2022; 14:eabl6992. [PMID: 35235340 DOI: 10.1126/scitranslmed.abl6992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SERAC1 deficiency is associated with the mitochondrial 3-methylglutaconic aciduria with deafness, (hepatopathy), encephalopathy, and Leigh-like disease [MEGD(H)EL] syndrome, but the role of SERAC1 in mitochondrial physiology remains unknown. Here, we generated Serac1-/- mice that mimic the major diagnostic clinical and biochemical phenotypes of the MEGD(H)EL syndrome. We found that SERAC1 localizes to the outer mitochondrial membrane and is a protein component of the one-carbon cycle. By interacting with the mitochondrial serine transporter protein SFXN1, SERAC1 facilitated and was required for SFXN1-mediated serine transport from the cytosol to the mitochondria. Loss of SERAC1 impaired the one-carbon cycle and disrupted the balance of the nucleotide pool, which led to primary mitochondrial DNA (mtDNA) depletion in mice, HEK293T cells, and patient-derived immortalized lymphocyte cells due to insufficient supply of nucleotides. Moreover, both in vitro and in vivo supplementation of nucleosides/nucleotides restored mtDNA content and mitochondrial function. Collectively, our findings suggest that MEGD(H)EL syndrome shares both clinical and molecular features with the mtDNA depletion syndrome, and nucleotide supplementation may be an effective therapeutic strategy for MEGD(H)EL syndrome.
Collapse
Affiliation(s)
- Hezhi Fang
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Anran Xie
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Miaomiao Du
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou 310000, China.,Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Xueyun Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China.,Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 318000, China
| | - Kaiqiang Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yinxu Fu
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiangshu Yuan
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Runxiao Fan
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Weidong Yu
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhuohua Zhou
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Tiantian Sang
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Ke Nie
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jin Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Qiongya Zhao
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou 310000, China
| | - Zhehui Chen
- Department of Pediatrics, Peking University First Hospital, Beijing 100000, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing 100000, China
| | - Chaoyang Hong
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| | - Jianxin Lyu
- Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou 325035, China.,School of Laboratory Medicine, Hangzhou Medical College, Hangzhou 310000, China.,Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou 310000, China
| |
Collapse
|
14
|
Zeng D, Guo X. Mantle Transcriptome Provides Insights into Biomineralization and Growth Regulation in the Eastern Oyster (Crassostrea virginica). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:82-96. [PMID: 34989931 DOI: 10.1007/s10126-021-10088-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Growth of the eastern oyster Crassostrea virginica, a major aquaculture species in the USA, is highly variable and not well understood at molecular levels. As growth of mollusks is confined in shells constructed by the mantle, mantle transcriptomes of large (fast-growing) and small (slow-growing) eastern oysters were sequenced and compared in this study. Transcription was observed for 31,186 genes, among which 104 genes were differentially expressed between the large and small oysters, including 48 upregulated and 56 downregulated in large oysters. Differentially expressed genes (DEGs) included genes from diverse pathways highlighting the complexity of shell formation and growth regulations. Seventeen of the 48 upregulated DEGs were related to shell matrix formation, most of which were upregulated in large oysters, indicating that large oysters are more active in biomineralization and shell formation. Genomic and transcriptomic analyses identified 22 genes encoding novel polyalanine containing proteins (Pacps) with characteristic motifs for matrix function that are tandemly duplicated on one chromosome, all specifically expressed in mantle and at higher levels in large oysters, suggesting that these expanded Pacps play important roles in shell formation and growth. Analysis of sequence variation identified 244,964 SNPs with 328 associated with growth. This study provides novel candidate genes and markers for shell formation and growth, and suggests that genes related to shell formation are important for the complex regulation of growth in the eastern oyster and possibly other bivalve mollusks. Results of this study show that both transcriptional modulation and functional polymorphism are important in determining growth.
Collapse
Affiliation(s)
- Dan Zeng
- Haskin Shellfish Research Laboratory, Department of Marine and Coastal Sciences, Rutgers University, 6959 Miller Avenue, Port Norris, New Jersey, 08349, USA
- College of Life and Environmental Science, Hunan University of Arts and Science, 3150 Dongting Road, Wuling District, Changde, Hunan, 415000, China
| | - Ximing Guo
- Haskin Shellfish Research Laboratory, Department of Marine and Coastal Sciences, Rutgers University, 6959 Miller Avenue, Port Norris, New Jersey, 08349, USA.
| |
Collapse
|
15
|
Molecular Genetics Overview of Primary Mitochondrial Myopathies. J Clin Med 2022; 11:jcm11030632. [PMID: 35160083 PMCID: PMC8836969 DOI: 10.3390/jcm11030632] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
Mitochondrial disorders are the most common inherited conditions, characterized by defects in oxidative phosphorylation and caused by mutations in nuclear or mitochondrial genes. Due to its high energy request, skeletal muscle is typically involved. According to the International Workshop of Experts in Mitochondrial Diseases held in Rome in 2016, the term Primary Mitochondrial Myopathy (PMM) should refer to those mitochondrial disorders affecting principally, but not exclusively, the skeletal muscle. The clinical presentation may include general isolated myopathy with muscle weakness, exercise intolerance, chronic ophthalmoplegia/ophthalmoparesis (cPEO) and eyelids ptosis, or multisystem conditions where there is a coexistence with extramuscular signs and symptoms. In recent years, new therapeutic targets have been identified leading to the launch of some promising clinical trials that have mainly focused on treating muscle symptoms and that require populations with defined genotype. Advantages in next-generation sequencing techniques have substantially improved diagnosis. So far, an increasing number of mutations have been identified as responsible for mitochondrial disorders. In this review, we focused on the principal molecular genetic alterations in PMM. Accordingly, we carried out a comprehensive review of the literature and briefly discussed the possible approaches which could guide the clinician to a genetic diagnosis.
Collapse
|
16
|
Chong M, Mohammadi-Shemirani P, Perrot N, Nelson W, Morton R, Narula S, Lali R, Khan I, Khan M, Judge C, Machipisa T, Cawte N, O'Donnell M, Pigeyre M, Akhabir L, Paré G. GWAS and ExWAS of blood mitochondrial DNA copy number identifies 71 loci and highlights a potential causal role in dementia. eLife 2022; 11:e70382. [PMID: 35023831 PMCID: PMC8865845 DOI: 10.7554/elife.70382] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/11/2022] [Indexed: 12/16/2022] Open
Abstract
Background Mitochondrial DNA copy number (mtDNA-CN) is an accessible blood-based measurement believed to capture underlying mitochondrial (MT) function. The specific biological processes underpinning its regulation, and whether those processes are causative for disease, is an area of active investigation. Methods We developed a novel method for array-based mtDNA-CN estimation suitable for biobank-scale studies, called 'automatic mitochondrial copy (AutoMitoC).' We applied AutoMitoC to 395,781 UKBiobank study participants and performed genome- and exome-wide association studies, identifying novel common and rare genetic determinants. Finally, we performed two-sample Mendelian randomization to assess whether genetically low mtDNA-CN influenced select MT phenotypes. Results Overall, genetic analyses identified 71 loci for mtDNA-CN, which implicated several genes involved in rare mtDNA depletion disorders, deoxynucleoside triphosphate (dNTP) metabolism, and the MT central dogma. Rare variant analysis identified SAMHD1 mutation carriers as having higher mtDNA-CN (beta = 0.23 SDs; 95% CI, 0.18-0.29; p=2.6 × 10-19), a potential therapeutic target for patients with mtDNA depletion disorders, but at increased risk of breast cancer (OR = 1.91; 95% CI, 1.52-2.40; p=2.7 × 10-8). Finally, Mendelian randomization analyses suggest a causal effect of low mtDNA-CN on dementia risk (OR = 1.94 per 1 SD decrease in mtDNA-CN; 95% CI, 1.55-2.32; p=7.5 × 10-4). Conclusions Altogether, our genetic findings indicate that mtDNA-CN is a complex biomarker reflecting specific MT processes related to mtDNA regulation, and that these processes are causally related to human diseases. Funding No funds supported this specific investigation. Awards and positions supporting authors include: Canadian Institutes of Health Research (CIHR) Frederick Banting and Charles Best Canada Graduate Scholarships Doctoral Award (MC, PM); CIHR Post-Doctoral Fellowship Award (RM); Wellcome Trust Grant number: 099313/B/12/A; Crasnow Travel Scholarship; Bongani Mayosi UCT-PHRI Scholarship 2019/2020 (TM); Wellcome Trust Health Research Board Irish Clinical Academic Training (ICAT) Programme Grant Number: 203930/B/16/Z (CJ); European Research Council COSIP Grant Number: 640580 (MO); E.J. Moran Campbell Internal Career Research Award (MP); CISCO Professorship in Integrated Health Systems and Canada Research Chair in Genetic and Molecular Epidemiology (GP).
Collapse
Affiliation(s)
- Michael Chong
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamiltonCanada
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
| | - Pedrum Mohammadi-Shemirani
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Thrombosis and Atherosclerosis Research InstituteHamiltonCanada
| | - Nicolas Perrot
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
| | - Walter Nelson
- Centre for Data Science and Digital Health, Hamilton Health SciencesHamiltonCanada
| | - Robert Morton
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
| | - Sukrit Narula
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Health Research Methods, Evidence, and Impact, McMaster UniversityHamiltonCanada
| | - Ricky Lali
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Health Research Methods, Evidence, and Impact, McMaster UniversityHamiltonCanada
| | - Irfan Khan
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
| | - Mohammad Khan
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Medicine, McMaster University, Michael G. DeGroote School of MedicineHamiltonCanada
| | - Conor Judge
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- National University of Ireland, GalwayGalwayIreland
| | - Tafadzwa Machipisa
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Medicine, University of Cape Town & Groote Schuur HospitalCape TownSouth Africa
- Hatter Institute for Cardiovascular Diseases Research in Africa (HICRA) & Cape Heart Institute (CHI), Department of Medicine, University of Cape TownCape TownSouth Africa
| | - Nathan Cawte
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
| | - Martin O'Donnell
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- National University of Ireland, GalwayGalwayIreland
| | - Marie Pigeyre
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Medicine, McMaster University, Michael G. DeGroote School of MedicineHamiltonCanada
| | - Loubna Akhabir
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Medicine, McMaster University, Michael G. DeGroote School of MedicineHamiltonCanada
| | - Guillaume Paré
- Department of Biochemistry and Biomedical Sciences, McMaster UniversityHamiltonCanada
- Department of Pathology and Molecular Medicine, McMaster UniversityHamiltonCanada
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health SciencesHamiltonCanada
- Department of Health Research Methods, Evidence, and Impact, McMaster UniversityHamiltonCanada
- Department of Medicine, McMaster University, Michael G. DeGroote School of MedicineHamiltonCanada
| |
Collapse
|
17
|
Mohamed A, Bakir T, Al-Hawel H, Al-Sharif I, Bakheet R, Kouser L, Murugaiah V, Al-Mozaini M. HIV-2 Vpx neutralizes host restriction factor SAMHD1 to promote viral pathogenesis. Sci Rep 2021; 11:20984. [PMID: 34697376 PMCID: PMC8545964 DOI: 10.1038/s41598-021-00415-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 10/12/2021] [Indexed: 11/21/2022] Open
Abstract
SAMHD1, a human host factor found in myeloid cells which restricts HIV-1 replication. It depletes the dNTPs pool for viral cDNA syntheses, thus preventing the viral replication in the cells. The viral accessory protein, Vpx, exists only in SIVmac/HIV-2 particles. Vpx in SIVmac can induce proteosomal degradation of SAMHD1, which then leads to a decrease in the cytoplasmic dNTP pool. The protein-protein interaction between Vpx and SAMHD1 and its consequences are still unclear. Methods: In this study, we cloned, for the first time, Vpx gene from a HIV-2 infected patient and found up to 30% sequence variation compared to known HIV-2 strains. We then analyzed the role of SAMHD1 protein expression in transfected THP-1 and U937 cells by transfecting with the Vpx gene derived from SIVmac, HIV-2 from the NIH sample as well as HIV-2 from a Saudi patient. We found that Vpx gene expression led to reduced levels of intracellular SAMHD1. When the supernatants of the transfected cell lines were examined for secreted cytokines, chemokines and growth factors, Vpx expression seemed to be suppressive of pro-inflammatory response, and skewed the immune response towards an anti-inflammatory response. These results suggest that Vpx can act at two levels: clearance of intracellular restriction factor and suppression of cytokine storm: both aimed at long-term latency and host-pathogen stand-off, suggesting that Vpx is likely to be a potential therapeutic target.
Collapse
Affiliation(s)
- Ahlam Mohamed
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, PO Box 3354 (MBC-03), Riyadh, 11211, Kingdom of Saudi Arabia
| | - Talal Bakir
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Huda Al-Hawel
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, PO Box 3354 (MBC-03), Riyadh, 11211, Kingdom of Saudi Arabia
| | - Ibtihaj Al-Sharif
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, PO Box 3354 (MBC-03), Riyadh, 11211, Kingdom of Saudi Arabia
| | - Razan Bakheet
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, PO Box 3354 (MBC-03), Riyadh, 11211, Kingdom of Saudi Arabia
| | | | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - Maha Al-Mozaini
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, PO Box 3354 (MBC-03), Riyadh, 11211, Kingdom of Saudi Arabia.
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
18
|
Yagüe-Capilla M, Castillo-Acosta VM, Bosch-Navarrete C, Ruiz-Pérez LM, González-Pacanowska D. A Mitochondrial Orthologue of the dNTP Triphosphohydrolase SAMHD1 Is Essential and Controls Pyrimidine Homeostasis in Trypanosoma brucei. ACS Infect Dis 2021; 7:318-332. [PMID: 33417760 DOI: 10.1021/acsinfecdis.0c00551] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The maintenance of deoxyribonucleotide triphosphate (dNTP) homeostasis through synthesis and degradation is critical for accurate genomic and mitochondrial DNA replication fidelity. Trypanosoma brucei makes use of both the salvage and de novo pathways for the provision of pyrimidine dNTPs. In this respect, the sterile α motif and histidine-aspartate domain-containing protein 1 (SAMHD1) appears to be the most relevant dNTPase controlling dNTP/deoxynucleoside homeostasis in mammalian cells. Here, we have characterized the role of a unique trypanosomal SAMHD1 orthologue denominated TbHD52. Our results show that TbHD52 is a mitochondrial enzyme essential in bloodstream forms of T. brucei. Knockout cells are pyrimidine auxotrophs that exhibit strong defects in genomic integrity, cell cycle progression, and nuclear DNA and kinetoplast segregation in the absence of extracellular thymidine. The lack of TbHD52 can be counteracted by the overexpression of human dCMP deaminase, an enzyme that is directly involved in dUMP formation yet absent in trypanosomes. Furthermore, the cellular dNTP quantification and metabolomic analysis of TbHD52 null mutants revealed perturbations in the nucleotide metabolism with a substantial accumulation of dCTP and cytosine-derived metabolites while dTTP formation was significantly reduced. We propose that this HD-domain-containing protein unique to kinetoplastids plays an essential role in pyrimidine dNTP homeostasis and contributes to the provision of deoxycytidine required for cellular dTTP biosynthesis.
Collapse
Affiliation(s)
- Miriam Yagüe-Capilla
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Armilla (Granada) 18016, Spain
| | - Víctor M. Castillo-Acosta
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Armilla (Granada) 18016, Spain
| | - Cristina Bosch-Navarrete
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Armilla (Granada) 18016, Spain
| | - Luis Miguel Ruiz-Pérez
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Armilla (Granada) 18016, Spain
| | - Dolores González-Pacanowska
- Instituto de Parasitología y Biomedicina “López-Neyra”, Consejo Superior de Investigaciones Científicas, Parque Tecnológico de Ciencias de la Salud, Armilla (Granada) 18016, Spain
| |
Collapse
|
19
|
Khan A, Sergi C. SAMHD1 as the Potential Link Between SARS-CoV-2 Infection and Neurological Complications. Front Neurol 2020; 11:562913. [PMID: 33101175 PMCID: PMC7546029 DOI: 10.3389/fneur.2020.562913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 08/24/2020] [Indexed: 12/18/2022] Open
Abstract
The recent pandemic of coronavirus infectious illness 2019 (COVID19) triggered by SARS-CoV-2 has rapidly spread around the globe, generating in severe events an acute, highly lethal pneumonia and death. In the past two hitherto similar CoVs, the severe acute respiratory syndrome CoV (SARS-CoV-1) and Middle East respiratory syndrome CoV (MERS-CoV) also gained universal attention as they produced clinical symptoms similar to those of SARS-CoV-2 utilizing angiotensin-converting enzyme 2 (ACE2) receptor and dipeptidyl peptidase 4 (DPP4) to go into the cells. COVID-19 may also present with overtly neurological symptoms. The proper understanding of the expression and dissemination of ACE2 in central and peripheral nerve systems is crucial to understand better the neurological morbidity caused by COVID-19. Using the STRING bioinformatic tool and references through text mining tools associated to Coronaviruses, we identified SAMHD1 as the probable link to neurological symptoms. Paralleled to the response to influenza A virus and, specifically, respiratory syncytial virus, SARS-CoV-2 evokes a response that needs robust induction of a subclass of cytokines, including the Type I and, obviously, Type III interferons as well as a few chemokines. We correlate ACE2 to the pathogenesis and neurologic complications of COVID-19 and found that SAMHD1 links to NF-κB pathway. No correlation was found with other molecules associated with Coronavirus infection, including ADAR, BST2, IRF3, IFITM3, ISG15, MX1, MX2, RNASEL, RSAD2, and VPRBP. We suggest that SAMHD1 is the molecule that may be behind the mechanisms of the neurological complications associated with COVID-19.
Collapse
Affiliation(s)
- Aiza Khan
- Department of Laboratory Medicine and Pathology, University of Albert Hospital, Edmonton, AB, Canada
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology, University of Albert Hospital, Edmonton, AB, Canada
- Department of Pediatrics, Stollery Children's Hospital, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
20
|
Davenne T, Klintman J, Sharma S, Rigby RE, Blest HTW, Cursi C, Bridgeman A, Dadonaite B, De Keersmaecker K, Hillmen P, Chabes A, Schuh A, Rehwinkel J. SAMHD1 Limits the Efficacy of Forodesine in Leukemia by Protecting Cells against the Cytotoxicity of dGTP. Cell Rep 2020; 31:107640. [PMID: 32402273 PMCID: PMC7225753 DOI: 10.1016/j.celrep.2020.107640] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 03/12/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
The anti-leukemia agent forodesine causes cytotoxic overload of intracellular deoxyguanosine triphosphate (dGTP) but is efficacious only in a subset of patients. We report that SAMHD1, a phosphohydrolase degrading deoxyribonucleoside triphosphate (dNTP), protects cells against the effects of dNTP imbalances. SAMHD1-deficient cells induce intrinsic apoptosis upon provision of deoxyribonucleosides, particularly deoxyguanosine (dG). Moreover, dG and forodesine act synergistically to kill cells lacking SAMHD1. Using mass cytometry, we find that these compounds kill SAMHD1-deficient malignant cells in patients with chronic lymphocytic leukemia (CLL). Normal cells and CLL cells from patients without SAMHD1 mutation are unaffected. We therefore propose to use forodesine as a precision medicine for leukemia, stratifying patients by SAMHD1 genotype or expression.
Collapse
Affiliation(s)
- Tamara Davenne
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK; Laboratory for Disease Mechanisms in Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Jenny Klintman
- Molecular Diagnostic Centre, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Sushma Sharma
- Department of Medical Biochemistry and Biophysics and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - Rachel E Rigby
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Henry T W Blest
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Chiara Cursi
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Anne Bridgeman
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Bernadeta Dadonaite
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Kim De Keersmaecker
- Laboratory for Disease Mechanisms in Cancer, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Peter Hillmen
- St James' Institute of Oncology, St James' University Hospital, Leeds LS9 7TF, UK
| | - Andrei Chabes
- Department of Medical Biochemistry and Biophysics and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 901 87 Umeå, Sweden
| | - Anna Schuh
- Molecular Diagnostic Centre, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; Department of Oncology, Old Road Campus Research Building, University of Oxford, Oxford OX3 7DQ, UK; Department of Haematology, Oxford University Hospitals NHS Trust, Oxford OX3 7JL, UK
| | - Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
21
|
Franzolin E, Coletta S, Ferraro P, Pontarin G, D'Aronco G, Stevanoni M, Palumbo E, Cagnin S, Bertoldi L, Feltrin E, Valle G, Russo A, Bianchi V, Rampazzo C. SAMHD1‐deficient fibroblasts from Aicardi‐Goutières Syndrome patients can escape senescence and accumulate mutations. FASEB J 2019; 34:631-647. [DOI: 10.1096/fj.201902508r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 01/16/2023]
Affiliation(s)
| | - Sara Coletta
- Department of Biology University of Padova Padova Italy
| | - Paola Ferraro
- Department of Biology University of Padova Padova Italy
| | | | | | | | - Elisa Palumbo
- Department of Molecular Medicine University of Padova Padova Italy
| | - Stefano Cagnin
- Department of Biology University of Padova Padova Italy
- CRIBI Biotechnology Center University of Padova Padova Italy
- CIR‐Myo Myology Center University of Padova Padova Italy
| | | | - Erika Feltrin
- Department of Biology University of Padova Padova Italy
| | - Giorgio Valle
- Department of Biology University of Padova Padova Italy
| | - Antonella Russo
- Department of Molecular Medicine University of Padova Padova Italy
| | - Vera Bianchi
- Department of Biology University of Padova Padova Italy
| | | |
Collapse
|
22
|
Interferon γ and α Have Differential Effects on SAMHD1, a Potent Antiviral Protein, in Feline Lymphocytes. Viruses 2019; 11:v11100921. [PMID: 31600877 PMCID: PMC6832628 DOI: 10.3390/v11100921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/05/2019] [Accepted: 10/06/2019] [Indexed: 12/15/2022] Open
Abstract
Sterile alpha motif and histidine/aspartic domain-containing protein 1 (SAMHD1) is a protein with anti-viral, anti-neoplastic, and anti-inflammatory properties. By degrading cellular dNTPs to constituent deoxynucleoside and free triphosphate, SAMHD1 limits viral DNA synthesis and prevents replication of HIV-1 and some DNA viruses such as HBV, vaccinia, and HSV-1. Recent findings suggest SAMHD1 is broadly active against retroviruses in addition to HIV-1, such as HIV-2, FIV, BIV, and EIAV. Interferons are cytokines produced by lymphocytes and other cells that induce a wide array of antiviral proteins, including some with activity again lentiviruses. Here we evaluated the role of IFNs on SAMHD1 gene expression, transcription, and post-translational modification in a feline CD4+ T cell line (FeTJ) and in primary feline CD4+ T lymphocytes. SAMHD1 mRNA in FetJ cells increased in a dose-related manner in response to IFNγ treatment concurrent with increased nuclear localization and phosphorylation. IFNα treatment induced SAMHD1 mRNA but did not significantly alter SAMHD1 protein detection, phosphorylation, or nuclear translocation. In purified primary feline CD4+ lymphocytes, IL2 supplementation increased SAMHD1 expression, but the addition of IFNγ did not further alter SAMHD1 protein expression or nuclear localization. Thus, the effect of IFNγ on SAMHD1 expression is cell-type dependent, with increased translocation to the nucleus and phosphorylation in FeTJ but not primary CD4+ lymphocytes. These findings imply that while SAMH1 is inducible by IFNγ, overall activity is cell type and compartment specific, which is likely relevant to the establishment of lentiviral reservoirs in quiescent lymphocyte populations.
Collapse
|
23
|
Asadian P, Finnie G, Bienzle D. The expression profile of sterile alpha motif and histidine-aspartate domain-containing protein 1 (SAMHD1) in feline tissues. Vet Immunol Immunopathol 2017; 195:7-18. [PMID: 29249320 DOI: 10.1016/j.vetimm.2017.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/29/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022]
Abstract
SAMHD1 restricts lentiviruses by limiting availability of deoxynucleoside triphosphates for reverse transcription. HIV-2 and SIV have virion-associated proteins to counteract SAMHD1. Cats have an ortholog to human SAMHD1 and the FIV is restricted by human SAMHD1, but expression of feline SAMHD1 is unknown. Using a whole-body tissue microarray consisting of 24 tissues for immunohistochemistry, SAMHD1 expression was identified in a wide range of cat tissues. SAMHD1 was most strongly expressed in skin and mucosal epithelium, and in hemolymphatic and spermatogenic tissues. Both nuclear and cytoplasmic expression was detected. Feline cell lines susceptible to FIV infection also highly expressed SAMHD1. Preferential expression of SAMHD1 at sites of viral entry and replication supports a role for feline SAMHD1 in restricting FIV.
Collapse
Affiliation(s)
- Peyman Asadian
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Gillian Finnie
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
24
|
Castellano P, Prevedel L, Eugenin EA. HIV-infected macrophages and microglia that survive acute infection become viral reservoirs by a mechanism involving Bim. Sci Rep 2017; 7:12866. [PMID: 28993666 PMCID: PMC5634422 DOI: 10.1038/s41598-017-12758-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 09/15/2017] [Indexed: 12/17/2022] Open
Abstract
While HIV kills most of the cells it infects, a small number of infected cells survive and become latent viral reservoirs, posing a significant barrier to HIV eradication. However, the mechanism by which immune cells resist HIV-induced apoptosis is still incompletely understood. Here, we demonstrate that while acute HIV infection of human microglia/macrophages results in massive apoptosis, a small population of HIV-infected cells survive infection, silence viral replication, and can reactivate viral production upon specific treatments. We also found that HIV fusion inhibitors intended for use as antiretroviral therapies extended the survival of HIV-infected macrophages. Analysis of the pro- and anti-apoptotic pathways indicated no significant changes in Bcl-2, Mcl-1, Bak, Bax or caspase activation, suggesting that HIV blocks a very early step of apoptosis. Interestingly, Bim, a highly pro-apoptotic negative regulator of Bcl-2, was upregulated and recruited into the mitochondria in latently HIV-infected macrophages both in vitro and in vivo. Together, these results demonstrate that macrophages/microglia act as HIV reservoirs and utilize a novel mechanism to prevent HIV-induced apoptosis. Furthermore, they also suggest that Bim recruitment to mitochondria could be used as a biomarker of viral reservoirs in vivo.
Collapse
Affiliation(s)
- Paul Castellano
- Public Health Research Institute (PHRI), Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of NJ, Newark, NJ, USA
| | - Lisa Prevedel
- Public Health Research Institute (PHRI), Newark, NJ, USA
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of NJ, Newark, NJ, USA
| | - Eliseo A Eugenin
- Public Health Research Institute (PHRI), Newark, NJ, USA.
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of NJ, Newark, NJ, USA.
| |
Collapse
|
25
|
Robledo D, Rubiolo JA, Cabaleiro S, Martínez P, Bouza C. Differential gene expression and SNP association between fast- and slow-growing turbot (Scophthalmus maximus). Sci Rep 2017; 7:12105. [PMID: 28935875 PMCID: PMC5608734 DOI: 10.1038/s41598-017-12459-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022] Open
Abstract
Growth is among the most important traits for animal breeding. Understanding the mechanisms underlying growth differences between individuals can contribute to improving growth rates through more efficient breeding schemes. Here, we report a transcriptomic study in muscle and brain of fast- and slow-growing turbot (Scophthalmus maximus), a relevant flatfish in European and Asian aquaculture. Gene expression and allelic association between the two groups were explored. Up-regulation of the anaerobic glycolytic pathway in the muscle of fast-growing fish was observed, indicating a higher metabolic rate of white muscle. Brain expression differences were smaller and not associated with major growth-related genes, but with regulation of feeding-related sensory pathways. Further, SNP variants showing frequency differences between fast- and slow-growing fish pointed to genomic regions likely involved in growth regulation, and three of them were individually validated through SNP typing. Although different mechanisms appear to explain growth differences among families, general mechanisms seem also to be involved, and thus, results provide a set of useful candidate genes and markers to be evaluated for more efficient growth breeding programs and to perform comparative genomic studies of growth in fish and vertebrates.
Collapse
Affiliation(s)
- Diego Robledo
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain.,The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, United Kingdom
| | - Juan A Rubiolo
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain
| | - Santiago Cabaleiro
- Cluster de Acuicultura de Galicia (Punta do Couso), Aguiño-Ribeira, 15695, Spain
| | - Paulino Martínez
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain
| | - Carmen Bouza
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain.
| |
Collapse
|
26
|
Leanza L, Romio M, Becker KA, Azzolini M, Trentin L, Managò A, Venturini E, Zaccagnino A, Mattarei A, Carraretto L, Urbani A, Kadow S, Biasutto L, Martini V, Severin F, Peruzzo R, Trimarco V, Egberts JH, Hauser C, Visentin A, Semenzato G, Kalthoff H, Zoratti M, Gulbins E, Paradisi C, Szabo I. Direct Pharmacological Targeting of a Mitochondrial Ion Channel Selectively Kills Tumor Cells In Vivo. Cancer Cell 2017; 31:516-531.e10. [PMID: 28399409 DOI: 10.1016/j.ccell.2017.03.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 02/03/2017] [Accepted: 03/07/2017] [Indexed: 12/13/2022]
Abstract
The potassium channel Kv1.3 is highly expressed in the mitochondria of various cancerous cells. Here we show that direct inhibition of Kv1.3 using two mitochondria-targeted inhibitors alters mitochondrial function and leads to reactive oxygen species (ROS)-mediated death of even chemoresistant cells independently of p53 status. These inhibitors killed 98% of ex vivo primary chronic B-lymphocytic leukemia tumor cells while sparing healthy B cells. In orthotopic mouse models of melanoma and pancreatic ductal adenocarcinoma, the compounds reduced tumor size by more than 90% and 60%, respectively, while sparing immune and cardiac functions. Our work provides direct evidence that specific pharmacological targeting of a mitochondrial potassium channel can lead to ROS-mediated selective apoptosis of cancer cells in vivo, without causing significant side effects.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Matteo Romio
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Michele Azzolini
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Antonella Managò
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Elisa Venturini
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Angela Zaccagnino
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Andrea Mattarei
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy
| | - Luca Carraretto
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Andrea Urbani
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Lucia Biasutto
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Veronica Martini
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Filippo Severin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Roberta Peruzzo
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy
| | - Valentina Trimarco
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Jan-Hendrik Egberts
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Charlotte Hauser
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Andrea Visentin
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Hematology and Immunological Branch, University of Padova, and Venetian Institute for Molecular Medicine (VIMM), via G. Orus 2, 35129 Padova, Italy
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, Medical Faculty, CAU, Kiel, and Department of Surgery, UKSH, Campus Kiel, Arnold-Heller-Strasse 3 (Haus 17), 24105 Kiel, Germany
| | - Mario Zoratti
- Department of Biomedical Sciences, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany; Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA.
| | - Cristina Paradisi
- Department of Chemical Sciences, University of Padova, via F. Marzolo 1, 35121 Padova, Italy.
| | - Ildiko Szabo
- Department of Biology, University of Padova, viale G. Colombo 3, 35121 Padova, Italy; CNR Institute of Neuroscience, viale G. Colombo 3, 35121 Padova, Italy.
| |
Collapse
|
27
|
Wang L. Mitochondrial purine and pyrimidine metabolism and beyond. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2017; 35:578-594. [PMID: 27906631 DOI: 10.1080/15257770.2015.1125001] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Carefully balanced deoxynucleoside triphosphate (dNTP) pools are essential for both nuclear and mitochondrial genome replication and repair. Two synthetic pathways operate in cells to produce dNTPs, e.g., the de novo and the salvage pathways. The key regulatory enzymes for de novo synthesis are ribonucleotide reductase (RNR) and thymidylate synthase (TS), and this process is considered to be cytosolic. The salvage pathway operates both in the cytosol (TK1 and dCK) and the mitochondria (TK2 and dGK). Mitochondrial dNTP pools are separated from the cytosolic ones owing to the double membrane structure of the mitochondria, and are formed by the salvage enzymes TK2 and dGK together with NMPKs and NDPK in postmitotic tissues, while in proliferating cells the mitochondrial dNTPs are mainly imported from the cytosol produced by the cytosolic pathways. Imbalanced mitochondrial dNTP pools lead to mtDNA depletion and/or deletions resulting in serious mitochondrial diseases. The mtDNA depletion syndrome is caused by deficiencies not only in enzymes in dNTP synthesis (TK2, dGK, p53R2, and TP) and mtDNA replication (mtDNA polymerase and twinkle helicase), but also in enzymes in other metabolic pathways such as SUCLA2 and SUCLG1, ABAT and MPV17. Basic questions are why defects in these enzymes affect dNTP synthesis and how important is mitochondrial nucleotide synthesis in the whole cell/organism perspective? This review will focus on recent studies on purine and pyrimidine metabolism, which have revealed several important links that connect mitochondrial nucleotide metabolism with amino acids, glucose, and fatty acid metabolism.
Collapse
Affiliation(s)
- Liya Wang
- a Department of Anatomy, Physiology and Biochemistry , Swedish University of Agricultural Sciences , Uppsala , Sweden
| |
Collapse
|
28
|
Rampazzo C, Tozzi MG, Dumontet C, Jordheim LP. The druggability of intracellular nucleotide-degrading enzymes. Cancer Chemother Pharmacol 2015; 77:883-93. [PMID: 26614508 DOI: 10.1007/s00280-015-2921-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/13/2015] [Indexed: 01/24/2023]
Abstract
Nucleotide metabolism is the target of a large number of anticancer drugs including antimetabolites and specific enzyme inhibitors. We review scientific findings that over the last 10-15 years have allowed the identification of several intracellular nucleotide-degrading enzymes as cancer drug targets, and discuss further potential therapeutic applications for Rcl, SAMHD1, MTH1 and cN-II. We believe that enzymes involved in nucleotide metabolism represent potent alternatives to conventional cancer chemotherapy targets.
Collapse
Affiliation(s)
- Chiara Rampazzo
- Department of Biology, University of Padova, 35131, Padua, Italy
| | - Maria Grazia Tozzi
- Department of Biology, Biochemistry Unit, University of Pisa, Pisa, Italy
| | - Charles Dumontet
- Université de Lyon, 69000, Lyon, France.,Université de Lyon 1, 69622, Lyon, France.,Université de Lyon 1, 69000, Lyon, France.,INSERM U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France.,Centre Léon Bérard, 69008, Lyon, France.,Hospices Civils de Lyon, 69000, Lyon, France
| | - Lars Petter Jordheim
- Université de Lyon, 69000, Lyon, France. .,Université de Lyon 1, 69622, Lyon, France. .,Université de Lyon 1, 69000, Lyon, France. .,INSERM U1052, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France. .,CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon, 69000, Lyon, France. .,Centre Léon Bérard, 69008, Lyon, France. .,Equipe Anticorps-Anticancer, INSERM U1052 - CNRS UMR 5286, Faculté Rockefeller, Centre de Recherche en Cancérologie de Lyon, 8 avenue Rockefeller, 69008, Lyon, France.
| |
Collapse
|